ABSTRACT
In this study, we investigated the effect of acetate, the most concentrated short-chain fatty acid (SCFA) in the gut and bloodstream, on the susceptibility of primary human CD4+ T cells to HIV-1 infection. We report that HIV-1 replication is increased in CD3/CD28-costimulated CD4+ T cells upon acetate treatment. This enhancing effect correlates with increased expression of the early activation marker CD69 and impaired class I/II histone deacetylase (HDAC) activity. In addition, acetate enhances acetylation of histones H3 and H4 and augments HIV-1 integration into the genome of CD4+ T cells. Thus, we propose that upon antigen presentation, acetate influences class I/II HDAC activity that transforms condensed chromatin into a more relaxed structure. This event leads to a higher level of viral integration and enhanced HIV-1 production. In line with previous studies showing reactivation of latent HIV-1 by SCFAs, we provide evidence that acetate can also increase the susceptibility of primary human CD4+ T cells to productive HIV-1 infection.
IMPORTANCE Alterations in the fecal microbiota and intestinal epithelial damage involved in the gastrointestinal disorder associated with HIV-1 infection result in microbial translocation that leads to disease progression and virus-related comorbidities. Indeed, notably via production of short-chain fatty acids, bacteria migrating from the lumen to the intestinal mucosa could influence HIV-1 replication by epigenetic regulatory mechanisms, such as histone acetylation. We demonstrate that acetate enhances virus production in primary human CD4+ T cells. Moreover, we report that acetate impairs class I/II histone deacetylase activity and increases integration of HIV-1 DNA into the host genome. Therefore, it can be postulated that bacterial metabolites such as acetate modulate HIV-1-mediated disease progression.
KEYWORDS: CD4+ T cells, HIV-1, histone acetylation, histone deacetylases, integration, provirus
INTRODUCTION
Short-chain fatty acids (SCFAs) have been reported to play key roles in the development of many conditions and diseases, including HIV-1 infection (1–7). These carboxylic acids, which contain an aliphatic tail of 6 carbon atoms or less, are mainly synthesized in the cecum and proximal colon by anaerobic commensal bacteria composing the gut microbiota via a dynamic process alternating between the glycolytic pathway, the pentose phosphate pathway, and the Wood-Ljungdahl pathway (8–18). It is known that gut bacteria can transform indigestible food components (e.g., fiber, polysaccharides, and oligosaccharides) into saturated aliphatic organic acids carrying one to six carbons, such as formate (C1), acetate (C2), propionate (C3), butyrate (C4), valerate (C5), and caproate (C6), through various metabolic pathways (8–10, 14, 17, 19–24). The three major SCFAs, acetate, propionate, and butyrate, are found in an approximate molar ratio of 60:25:15 in the gut (8). This ratio, as well as the overall production of SCFAs, is influenced by many factors, including microbiota diversity, substrate source, and gut transit time. However, independently of this, it was established that acetate always remains the most concentrated SCFA in the colon and is the most important one found in the systemic circulation (25). Active transport mechanisms mediated by MCT-1 and SMCT-1 receptors allow SCFAs to reach the peripheral circulation via the hepatic and portal venous systems (8, 9, 26–28). The capacity of SCFAs to modulate and regulate numerous cell functions, such as ion transport, proliferation, differentiation, and gene expression, is initiated by their potential to ligate different surface receptors (10, 15). It has been demonstrated that the G protein-coupled receptors (GPCRs) FFAR2 (also called GPR43) and FFAR3 (also called GPR41) mediate signal transduction following their binding with different SCFAs (29). Like, all GPCRs, FFAR2 (which preferentially binds acetate and propionate) and FFAR3 (which binds to propionate, butyrate, and valerate) mainly signal via heterotrimeric G proteins (8, 30–33). In addition to these, MCT-1 or SMCT-1 transporters are also implicated in SCFA-mediated signal transduction events in many cell types, such as in diverse lymphocyte populations (34). In contrast to GPCRs, which respond to SCFAs through G-protein signal transduction cascades, these receptors promote SCFA uptake into the cell to modulate biological responses (9, 34). Once they have reached the cytoplasm, SCFAs are known to directly regulate gene expression by competitively inhibiting histone deacetylases (HDACs) at the hydrophobic cleft of their active site (35–37).
In the past few years, several groups have demonstrated that HDACs prevent gene transcription by removing histone-bound acetyl groups, a process leading to a closed and repressed chromatin structure (2, 38–40). Inhibitors of class I/II HDAC activity (such as SCFAs) block this epigenetic regulation and are involved in the development of different diseases. Recent publications have highlighted the role of HDACs in the repression of HIV-1 regulatory elements also called long-terminal repeats (LTR). In this case, negative transcription factors, such as Yin-Yang-1 (YY-1), activator protein-4 (AP-4), and the Sp1-myc complex, have been shown to recruit HDAC-1 to LTR or the TATA box and to restrain transcription from the viral promoter to maintain viral latency (2, 41–45). Interestingly, it has been reported that butyrate-producing bacteria residing in the mouth, vaginal, and gut mucous membranes are implicated in AIDS progression by inducing reactivation of HIV-1 proviral DNA in latently infected cell lines and mononuclear cells via inhibition of HDAC activity (2, 3, 6, 7, 36, 38, 41, 45–48).
Given the inherent ability of SCFAs to affect diverse biological cell functions and as HIV-1 infection leads to profound and early alterations of the permeability of the gut epithelium, we assessed the overall impact of acetate on the susceptibility of primary human CD4+ T cells to productive HIV-1 infection. Our studies were focused on acetate based on the idea that it is the most concentrated SCFA in the gut. Our results suggest that acetate treatment, when combined with CD3/CD28 costimulation to mimic antigen presentation, reduces class I/II HDAC activity, increases histone acetylation, and enhances HIV-1 integration into the host chromosomal DNA. It can thus be proposed that bacterial metabolites, such as acetate, could exert an influence on HIV-1-mediated disease progression.
RESULTS
Acetate increases HIV-1 replication in CD3/CD28-costimulated CD4+ T cells.
In the last few years, several studies have monitored the impact of SCFAs produced by bacteria on HIV-1 biology with particular emphasis on reactivation of virus gene expression (2–7, 49). Here, to characterize the potential modulatory effect of acetate on HIV-1 replication, quiescent primary human CD4+ T cells were purified by magnetic separation from peripheral blood mononuclear cells (PBMCs) from different healthy donors and were subjected to anti-CD3/CD28 costimulation in the absence or presence of acetate. The cells were next inoculated with replication-competent NL4.3Balenv virus (i.e., NL4.3 carrying Env glycoproteins of the R5-using Bal isolate), and the extracellular p24 contents were quantified either at the peak of virus production (i.e., 5 days postinfection [dpi]) or at various time points postinfection. The results displayed in Fig. 1A show that acetate enhances virus production in CD3/CD28-costimulated primary human CD4+ T cells. There was no virus replication in acetate-treated primary human CD4+ T cells, which were not subjected to anti-CD3/CD28 costimulation (data not shown). Experiments were also performed with the infectious reporter virus NL4.3Bal-IRES-HSA to measure the effect of acetate on HIV-1 replication at the single-cell level. An acetate-mediated enhancement in the percentage of heat-stable antigen (HSA)-expressing cells (indicative of the number of cells productively infected with HIV-1) was seen at 3 dpi (Fig. 1B). Our data also demonstrate that acetate augments the percentage of HIV-1-infected (i.e., HSA+) cells and not the mean fluorescence intensity (MFI) per individual cell, thus suggesting that acetate increases the overall sensitivity of primary human CD4+ T cells to productive HIV-1 infection without affecting virus gene expression at the single-cell level. Representative fluorescence-activated cell sorter (FACS) analyses are shown in Fig. 1C. We carried out experiments with increasing concentrations of acetate ranging from 2 to 40 mM. Cell viability was not affected by the acetate concentrations studied (Fig. 2A), and we detected an acetate-dependent, dose-dependent increase in HIV-1 infection (as monitored by estimating the percentage of HSA+ cells) (Fig. 2B). We selected the 20 mM concentration of acetate for the next series of experiments based on previous studies indicating that this level of acetate can be reached in the proximal and distal human colon (12). Together, these results suggest that acetate promotes productive HIV-1 infection in CD3/CD28-costimulated primary human CD4+ T cells.
FIG 1.
HIV-1 replication in CD3/CD28-costimulated CD4+ T cells is augmented by acetate. Purified primary human CD4+ T cells were costimulated with anti-CD3 and anti-CD28 MAbs in the absence or presence of acetate (20 mM). (A) The cells were next inoculated with replication-competent NL4.3Balenv viruses, and virus production was estimated either at 6 dpi (left) (6 donors are shown) or at the indicated time points postinfection (right) (a representative donor is shown) by measuring the p24 contents in cell-free supernatants. (B) In some experiments, untreated and acetate-treated cells were incubated with the NL4.3Bal-IRES-HSA reporter virus for 3 days before assessing HSA expression by flow cytometry. Percentages of HSA+ cells are depicted on the left, while the MFI is shown on the right. Each symbol represents a different donor, with the horizontal lines depicting the means of six donors tested. Statistical analyses were done using ratio-paired Student's t test. The asterisks denote statistically significant data (**, P ≤ 0.01; ***, P ≤ 0.001). (C) The gating strategy used in flow cytometry analyses to estimate the percentage of cells productively infected with HIV-1 (HSA+ as defined with an allophycocyanin [APC]-conjugated anti-HSA MAb) for each experimental condition consisted of live lymphocyte gating based on size and complexity on a forward scatter (FSC)/side scatter (SSC) plot (left), followed by doublet discrimination on an FSC-height (H)/FSC-width (W) plot (center), to finally gate HSA+ cells on an FSC-H/APC plot (right). Mock-infected cells were used as negative controls for HSA staining. The number in the plots indicates the percentage of cells within the gate.
FIG 2.

Acetate does not affect cell viability but induces a dose-dependent increase in HIV-1 replication. Purified primary human CD4+ T cells were costimulated with anti-CD3 and anti-CD28 MAbs in the absence or presence of increasing concentrations of acetate. (A) Cell viability was monitored by flow cytometry at day 6 following acetate treatment. (B) Purified primary human CD4+ T cells were first treated as described for panel A and then incubated with the NL4.3Bal-IRES-HSA reporter virus for 3 days before quantifying the percentages of HSA+ cells by flow cytometry. Each symbol represents a different donor, with the horizontal lines depicting the means of five donors tested. Statistical analyses were done using ratio-paired Student's t tests. The asterisks denote statistically significant data (*, P ≤ 0.05; **, P ≤ 0.01; ***, P ≤ 0.001).
Cell proliferation and activation profiles are affected differently by acetate treatment.
Cellular proliferation and activation are known to have an impact on the susceptibility of CD4+ T cells to HIV-1 infection. For example, cellular proliferation plays an important role in the HIV-1 life cycle by promoting virus dissemination, which helps to maintain viral reservoirs (50, 51), whereas cell activation allows the translocation of host transcription factors to the nucleus, where they trigger genes implicated in immune response and virus production (52, 53). Thus, cell proliferation was evaluated by the use of a dilution assay that is based on the fluorescent cell staining dye carboxyfluorescein succinimidyl ester (CFSE). We also studied the cell activation status by measuring the surface expression of some activation markers (i.e., CD25, CD69, and CD154) by flow cytometry. As expected, cell proliferation was induced in a statistically significant manner upon CD3/CD28 costimulation at the two time points tested (Fig. 3A). However, proliferation of CD4+ T cells was significantly decreased at the earliest time point by acetate treatment. Surface expression of the activation-associated receptors CD25, CD69, and CD154 was significantly induced following CD3/CD28 costimulation compared to untreated CD4+ T cells, while CD69 expression was the only surface marker to be further increased upon treatment with acetate (Fig. 3B). These observations demonstrate that acetate exhibits differential effects with respect to cell proliferation and activation.
FIG 3.
Acetate exerts differential effects on cell proliferation and activation markers. Purified primary human CD4+ T cells were first treated as described in the legend to Fig. 1. (A) A CFSE-based dilution assay was performed by flow cytometry to evaluate cell proliferation following acetate treatment for 3 or 6 days. Representative proliferation profiles are depicted on the left, whereas division indices are shown on the right. (B) Surface expression of some T cell activation markers (CD25, CD69, and CD154) was evaluated by flow cytometry following acetate treatment for 3 days. The data shown were obtained from CD4+ T cell preparations isolated from the peripheral blood of 4 (A) or 6 (B) distinct healthy donors. Each symbol represents a different donor, and the horizontal lines depict the means of all donors tested. Statistical analyses were done using one-way ANOVA, followed by a Dunnett multiple-comparison test. The asterisks denote statistically significant data (*, P ≤ 0.05; **, P ≤ 0.01; ****, P ≤ 0.0001).
Acetate does not modulate surface levels of CD4 but decreases CCR5 expression and virus entry into CD3/CD28-costimulated CD4+ T cells.
We aimed at elucidating the mechanism by which acetate promotes HIV-1 replication in CD3/CD28-costimulated primary human CD4+ T cells. To this end, we analyzed its impact on the surface expression of HIV-1 receptor and coreceptor, CD4 and CCR5. Although surface expression of both CD4 and CCR5 are significantly enhanced upon CD3/CD28 costimulation, acetate treatment does not alter surface levels of CD4 (Fig. 4A) but reduces CCR5 expression (Fig. 4B). Therefore, the acetate-mediated increase in HIV-1 production is not due to higher expression of cell surface receptors known to directly participate in the early events in the virus life cycle, such as attachment and/or entry. We performed a virus entry test to define whether the decrease in CCR5 surface levels affects the early steps in the HIV-1 life cycle. The results depicted in Fig. 5 demonstrate that HIV-1 entry was slightly lower in acetate-treated target cells.
FIG 4.
CCR5 surface expression in CD3/CD28-costimulated CD4+ T cells is diminished by acetate. Purified primary human CD4+ T cells were treated as specified in the legend to Fig. 1. Surface expression of CD4 (A) and CCR5 (B) in the total CD4+ T cell population was evaluated by flow cytometry. The data shown were obtained from CD4+ T cell preparations isolated from the peripheral blood of 5 (A) and 4 (B) distinct healthy donors. Each symbol represents a different donor, and the horizontal lines depict the means of all the donors tested. Statistical analyses were done using one-way ANOVA, followed by a Dunnett multiple-comparison test. The asterisks denote statistically significant data (*, P ≤ 0.05; **, P ≤ 0.01).
FIG 5.
Acetate decreases virus entry into CD3/CD28-costimulated CD4+ T cells. Purified primary human CD4+ T cells were subjected to CD3/CD28 costimulation in the absence or presence of acetate. Virus entry was estimated by quantifying the intracellular p24 contents by an enzyme-linked immunosorbent assay (ELISA) in the total cell population as described in Materials and Methods. The data shown represent the means and standard errors of the mean (SEM) of triplicate samples. Statistical analysis was performed using ratio-paired t tests (*, P < 0.05).
Acetate decreases class I/II HDAC activity in CD3/CD28-costimulated CD4+ T cells.
It is now well established that SCFAs, at physiological levels, can inhibit HDAC activity. Indeed, many groups have recently revealed the dominant role of this epigenetic regulatory mechanism in different contexts, such as neutrophil apoptosis and human colon cancer (40, 54, 55). Moreover, several publications have demonstrated the positive effect of butyrate on HIV-1 gene expression. Those data suggested that this SCFA promotes chromatin modification via histone acetylation and leads to HIV-1 reactivation in latently infected cells (2–7, 41, 46, 56). Based on the hypothesis that other SCFAs may induce epigenetic regulatory mechanisms such as histone acetylation to modulate HIV-1 replication, we investigated the ability of acetate to also act as a class I/II HDAC inhibitor in CD3/CD28-costimulated primary human CD4+ T cells. We found that acetate decreases class I/II HDAC activity in CD3/CD28-costimulated primary human CD4+ T cells (Fig. 6). As expected, the antifungal antibiotic trichostatin A (TSA), which is known to selectively inhibit class I/II mammalian HDACs, completely abrogates class I/II HDAC activity.
FIG 6.
Class I/II HDAC activity in CD3/CD28-costimulated CD4+ T cells is reduced by acetate. Purified primary human CD4+ T cells were treated as specified in the legend to Fig. 1. Some cells were also subjected to CD3/CD28 costimulation before treatment with TSA. The relative activity of class I/II HDACs in the total CD4+ T cell population was measured with the HDAC-Glo I/II assay according to the manufacturer's instructions. Each symbol represents a different donor, and the horizontal lines depict the means of 5 donors tested. Statistical analyses were done using one-way ANOVA, followed by a Dunnett multiple-comparison test. The asterisks denote statistically significant data (*, P ≤ 0.05).
Acetate induces acetylation of histones H3 and H4 in CD3/CD28-costimulated CD4+ T cells.
To establish a relationship between the acetate-mediated enhancement of HIV-1 infection in primary human CD4+ T cells and its HDAC-inhibitory activity, we performed intracellular flow cytometry analyses to quantify histone lysine acetylation events. The results from our studies reveal that acetate causes acetylation of both histones H3 (K9) and H4 (K5/8/12/16) in primary human CD4+ T cells (Fig. 7).
FIG 7.
Acetate mediates histone H3 and H4 acetylation. Primary human CD4+ T cells were first subjected to CD3/CD28 costimulation for 72 h in the absence or presence of acetate. Control cells were further stimulated for 4 h with the HDAC inhibitor TSA. Histone acetylation was detected by flow cytometry using MAbs specific for H3K9 (A) and H4 K5/8/12/16 (B). Each bar represents the mean percentages and standard deviations (SD) of positive cells from 5 distinct donors. Flow cytometry data obtained with cells costimulated with CD3 and CD28 MAbs were set at 100%. Statistical analyses were performed using ratio-paired t tests. The asterisks denote statistically significant data (*, P ≤ 0.05; **, P ≤ 0.01).
Acetate enhances integration of HIV-1 DNA in CD3/CD28-costimulated CD4+ T cells.
With its capacity to open chromatin by inhibiting class I/II HDAC activity, acetate could augment HIV-1 infection by facilitating provirus integration into the host genome (57, 58). To validate this hypothesis, we performed a combined Alu-HIV-1 PCR (where Alu-HIV-1 refers to Alu elements [a short stretch of DNA originally characterized by the action of the Arthrobacter luteus/Alu restriction endonuclease] and adjacent LTR/gag sequences [M667/M661]) and quantitative real-time PCR assay on purified primary CD4+ T cells costimulated with anti-CD3 and anti-CD28 monoclonal antibodies (MAbs) and either left untreated or treated with acetate. Moreover, we also measured HIV-1 completed reverse transcripts to further refine the mechanism of action of acetate (i.e., its modulatory effect on reverse transcription or integration). To discriminate between an effect of acetate on virus integration at the single-cell level versus an influence on cell-to-cell propagation, virus infection was allowed to proceed for 24 h only. Acetate caused a more significant increase in the number of copies of proviral DNA compared to the number of completed reverse transcripts (i.e., 1.57- versus 4.82-fold increase) (Fig. 8).
FIG 8.

Acetate exerts a more potent effect on HIV-1 integration events. Primary human CD4+ T cells from 6 distinct donors were first subjected to CD3/CD28 costimulation for 72 h in the absence or presence of acetate. Next, the cells were infected for 24 h with NL4.3Balenv viruses. The cells were processed to determine the number of copies of completed reverse transcripts (A) and proviral DNA (B) by a quantitative real-time PCR test. Each symbol represents a different donor, and the horizontal lines depict the means of 6 donors tested. Statistical analyses were done using ratio-paired Student's t tests. The asterisks denote statistically significant data (*, P ≤ 0.05; **, P ≤ 0.01).
DISCUSSION
Resulting from the fermentation of indigestible food components by anaerobic commensal bacteria composing the gut microbiome (8–10, 14, 17, 19–24), SCFAs have been suggested to affect HIV-1-mediated disease progression (1–7, 42). Previous studies have focused on the impact of the SCFA butyrate on reactivation of the provirus in cells latently infected with HIV-1. It has been suggested that butyrate acts as an inhibitor of class I HDACs, leading to histone hyperacetylation and enhancement of HIV-1 gene transcription (2, 3). Indeed, several lines of evidence indicate that HDACs repress HIV-1 LTR activity, whereas their inhibition increases the expression of viral genes (43–45). As enteric bacteria composing the gut microbiota are known to generate SCFAs as products of degradation of dietary fibers and these microorganisms are rapidly translocated to mucosal tissues following HIV-1 infection (59–64), the presence of these SCFAs could impact the overall permissiveness of CD4+ T cells to virus infection. The present work thus focused on acetate, the SCFA with the highest concentration in the colon and the major one found in the systemic circulation (8, 26). For instance, total acetate levels in humans range from a concentration of 63.4 mmol/kg of body weight in the ascending large intestine to 50 mmol/kg in the sigmoid/rectum anatomic segment (65). Moreover, the molar ratios of acetate to proprionate to butyrate are 91:5:4 in the peripheral blood (65). Importantly, our work was performed with a physiological concentration of acetate (i.e., 20 mM) based on previous studies indicating that the total concentrations of SCFAs vary from 70 to 140 mM in the proximal colon to 20 to 70 mM in the distal colon (12). Moreover, acetate, proprionate, and butyrate are found in an approximate molar ratio of 60:20:20 in the colon and stool (25, 66, 67).
First, we explored the impact of acetate on the replication of two R5-tropic NL4.3-based viruses in CD3/CD28-costimulated primary human CD4+ T cells. In contrast to previous studies, which focused on the effect of butyrate on reactivation of HIV-1 in latently infected cells (6), the present results underscore the impact of acetate exposure on the permissiveness of primary human CD4+ T cells to productive HIV-1 infection. Our observations revealed that acetate enhances the susceptibility of primary human CD4+ T cells to productive HIV-1 infection (i.e., a greater proportion of HIV-1-infected cells) without affecting virus gene expression at the single-cell level (Fig. 1 and 2).
It is now well established that CD4+ T cell activation is essential for the efficient replication of HIV-1 in these cells (68). While it is not required for viral entry, cellular activation is important for subsequent steps of the viral cycle, such as reverse transcription, integration of the provirus into the cell genome, and transcription of the viral genome (69). The influence of acetate treatment on cell activation was evaluated by assessing the expression of known surface activation markers (i.e., CD69, CD25, and CD154) by flow cytometry. As shown in Fig. 3B, the presence of acetate increases the expression of the early activation marker CD69 while surface expression of CD25 and CD154 are not affected. As CD25 and CD154 are, respectively, intermediate and late activation markers (70), it is possible that a stimulation period of 3 days is too brief to significantly modulate CD25 and CD154 among all the donors tested. Nonetheless, the increased expression of CD69 implies a positive effect of acetate on the activation profile of CD3/CD28-costimulated CD4+ T cells. As an enhanced activation state suggests greater susceptibility of CD4+ T cells to HIV-1 infection, acetate treatment could possibly promote HIV-1 infection via this mechanism.
Furthermore, it is now well established that cellular activation leads to a variety of responses in CD4+ T cells, such as their proliferation (71). Proliferation of infected cells increases their pool and contributes to the maintenance of reservoir cells (50, 51). As acetate treatment modulates the early activation marker CD69, we performed cell proliferation studies. Surprisingly, acetate treatment significantly decreased the average number of cell divisions at day 3 poststimulation. However, the acetate-mediated reduction in cell proliferation was no longer seen at a later time point (i.e., 6 days poststimulation). Considering the important role of the K+ channels in controlling T-cell proliferation (72–75) and that butyrate has been shown to downregulate calcium-activated K+ channels in human airway epithelial cells (76), it is possible that the observed delay of cellular proliferation could be partially caused by a similar effect of acetate on K+ channels in CD3/CD28-costimulated CD4+ T cells. Thus, despite having a positive impact on the cell surface activation marker profile, acetate delays cellular proliferation. Enhanced proliferation of virus-infected CD4+ T cells therefore cannot explain the effect of acetate on the greater permissiveness of CD3/CD28-costimulated CD4+ T cells to HIV-1 infection.
HIV-1 interacts with different constituents at the surfaces of its host cells. Two of them, the primary receptor CD4 and the predominant coreceptor CCR5, act sequentially in concert with the viral envelope protein gp120 to promote fusion of viral and cellular membranes and allow viral entry (77). As modulation of their expression could affect cellular permissiveness to HIV-1, we investigated the impact of acetate treatment on the expression of these two proteins at the surfaces of CD3/CD28-costimulated CD4+ T cells. As illustrated in Fig. 4, CD4 expression was significantly increased following CD3/CD28 costimulation but was not affected further by acetate treatment. However, while CCR5 expression was also significantly augmented at the surfaces of CD3/CD28-costimulated CD4+ T cells, concomitant exposure to acetate returned CCR5 surface expression to basal levels, near those found on the surfaces of quiescent cells. The influence of acetate on CCR5 expression is reminiscent of the action of ITF2357, another HDAC inhibitor, which was shown to inhibit the transcription of the CCR5 gene in PBMCs (78). As cell surface expression of CCR5 is correlated with the susceptibility of target cells to R5-tropic HIV-1 infection (79–81), our observations indicating that the process of virus entry is reduced by acetate are not unexpected (Fig. 5).
The mechanism by which acetate mediates a positive effect on HIV-1 replication in activated CD4+ T cells despite its capacity to decrease virus entry appears to be a paradox. Following multiple lines of studies demonstrating that HDAC inhibitors can drive HIV-1 gene expression in various experimental model systems of HIV-1 latency, we assessed the capacity of acetate to abrogate HDAC activity in CD3/CD28-costimulated primary human CD4+ T cells. We found that acetate decreased class I/II HDAC activity in such cells. Indeed, CD3/CD28 costimulation significantly increased class I/II HDAC activity at day 3 poststimulation, whereas treatment with acetate reduced HDAC activity to levels almost similar to those found in quiescent cells (Fig. 6). Importantly, we provide evidence that acetate induces acetylation of both histones H3 and H4 in primary human CD4+ T cells (Fig. 7). These results are highly significant considering that acetylation of the lysine residues at the N termini of histone proteins (primarily histones H3 and H4) results in decondensation of heterochromatin (i.e., the transcriptionally silent form of chromatin) into the more relaxed euchromatin structure (i.e., the transcriptionally active form of chromatin). Moreover, it is now established that HIV-1 preferentially integrates into the euchromatin based on in vitro (82, 83) and in vivo studies (83, 84).
Considering that inhibition of such chromatin modifiers facilitates a switch from repressive heterochromatin to permissive euchromatin (2), inhibition of class I/II HDAC activity could promote the integration of proviral DNA into host euchromatic domains. The results from HIV-1 integration assays, which combine an Alu-HIV-1 PCR and a quantitative real-time PCR test (85), showed that acetate can potently increase integration of HIV-1 DNA into the host's genome (Fig. 8). Interestingly, acetate displays a more significant impact on virus integration than completion of reverse transcripts. These results suggest that acetate, by acting as a class I/II HDAC inhibitor, could relieve epigenetic controls restricting retroviral integration into the host DNA, possibly by allowing the provirus to access open and active regions of genomic DNA where viral integration is strongly favored (86, 87).
Based on data described in the present study, we hypothesize that the bacterial metabolite acetate could play a role in the early stages of HIV-1 infection, such as initial spreading and reservoir establishment, by enhancing cellular activation and viral integration events. Considering that other SCFAs also act as inhibitors of class I/II HDAC activity (7, 35–37), their effect could be compounded by the action of acetate in a physiological setting. Therefore, further studies are required to provide a better understanding of the putative impact of bacterial metabolites such as SCFAs during HIV-1 pathogenesis.
MATERIALS AND METHODS
Ethics statement and cell culture.
The current study was approved by the Bioethics Committee of the Centre Hospitalier Universitaire de Québec-Université Laval. Samples were collected from healthy donors in accordance with the guidelines of the Institutional Bioethics Committee. All the donors provided written informed consent. To obtain purified primary human CD4+ T cells, PBMCs were isolated by Ficoll-Hypaque gradient centrifugation from healthy donors and kept frozen at −80°C at a final concentration of 100 × 106 cells/ml in freezing medium (90% fetal bovine serum [FBS] and 10% dimethyl sulfoxide [DMSO]) for a maximum of 3 months. Purified CD4+ T cells were isolated from thawed PBMCs by an immunomagnetic negative-selection procedure according to the manufacturer's instructions (StemCell Technologies, Vancouver, BC, Canada) and cultured in Roswell Park Memorial Institute (RPMI) 1640 medium (Thermo Fisher Scientific, Waltham, MA) supplemented with 10% FBS (Thermo Fisher Scientific) at a final concentration of 2 × 106 cells/ml.
Human embryonic kidney (HEK) 293T cells were kindly provided by Warner C. Greene (The J. Gladstone Institutes, San Francisco, CA), and TZM-bl indicator cells were obtained from John C. Kappes, Xiaoyun Wu, and Tranzyme Inc. via the AIDS Research Reagent Program (Germantown, MD) (88–92). The cell lines were cultured in Dulbecco's modified Eagle medium (DMEM) (Thermo Fisher Scientific) supplemented with 10% FBS.
Antibodies, reagents, and infectious virus constructs.
Allophycocyanin (APC)-conjugated anti-mouse CD24 (also known as HSA) was purchased from eBioscience (San Diego, CA). Fluorescein isothiocyanate (FITC)-conjugated anti-human CD4, APC-Cy7-conjugated anti-human CD195 (CCR5), phycoerythrin (PE)-conjugated anti-human CD25, FITC-conjugated anti-human CD69, and FITC-conjugated anti-human CD154 were all obtained from BD Biosciences (Mississauga, ON, Canada). The CellTrace CFSE Cell proliferation kit was purchased from Thermo Fisher Scientific. Hybridomas producing 183-H12-5C and 31-90-25 MAbs, which recognize different epitopes of the HIV-1 major viral core protein p24 Gag, were supplied by the AIDS Research Reagent Program and the ATCC (Manassas, VA), respectively. The rabbit anti-acetyl-histone H3 (K9) MAb (Alexa Fluor488 conjugate) was purchased from Cell Signaling Technology (Danvers, MA), while the anti-acetyl-histone H4 MAb (K5/8/12/16) was purchased from EMD Millipore (Etobicoke, ON, Canada). Mix-n'-Stain kit CF405M was purchased from Sigma-Aldrich (Saint Louis, MO) and used to stain anti-acetyl-histone H4 MAb following the manufacturer's protocol. The optimal concentration of the anti-acetyl-histone H4 MAb was determined by titration on TSA-stimulated and control cells before experiments were performed. A 200 mM stock solution of acetate was made by diluting glacial acetic acid (99.8% purity; Merck, Germany) in phosphate-buffered saline (PBS) and adjusted to a pH of 7.4 using NaOH. This neutral acetate solution was then filtered on a 0.22-μm syringe filter. Controls for acetate consisted of sterile PBS (pH 7.4). The infectious molecular vectors pNL4.3Balenv and pNL4.3Bal-IRES-HSA are described below.
Production of virus stocks.
The NL4.3Balenv vector codes for R5-tropic Bal envelope (Env) glycoproteins inserted within the NL4.3 backbone and generates fully infectious HIV-1 particles (93). The NL4.3Bal-IRES-HSA plasmid produces R5-using replication-competent reporter viruses and carries all the viral genes, as well as a reporter gene coding for the murine HSA protein, which is expressed on the membranes of productively infected cells via a glycosylphosphatidylinositol anchor (94). Virus preparations were made by calcium phosphate transfection of the infectious molecular clones pNL4.3Balenv and pNL4.3Bal-IRES-HSA. Briefly, HEK293T cells were transfected with the expression vectors, and cell-free supernatants were collected after 48 h. Newly produced viral particles were obtained following an ultracentrifugation step (100,000 × g for 45 min). The pellet was suspended in 2 ml of endotoxin-free PBS and frozen at −80°C until needed. Quantification of infectious virus titers harvested after ultracentrifugation was performed according to the Spearman-Karber method and following infection of TZM-bl indicator cells.
Stimulation and virus infection assays.
Purified primary human CD4+ T cells (1 × 106 cells per experimental condition) were cultured in RPMI 1640 medium supplemented with 10% FBS at a final concentration of 2 × 106 cells/ml, seeded in 48-well flat-bottom tissue culture plates, and either left untreated or treated with anti-CD3 (clone OKT3; 5 μg/ml) and anti-CD28 (clone 9.3; 2 μg/ml) MAbs in the presence or absence of acetate (at a final concentration of 20 mM for most experiments). After 72 h of incubation at 37°C under a 5% CO2 atmosphere, the cells were either left uninfected to analyze surface expression of CD4, CCR5, CD25, CD69, and CD154 by flow cytometry or inoculated with HIV-1. In some studies, cellular viability was assessed by flow cytometry using the fixable viability dye eFluor 780 (Thermo Fisher Scientific). Virus infection experiments were performed using NL4.3Balenv and NL4.3Bal-IRES-HSA virus preparations added to cells at a final multiplicity of infection (MOI) of 0.1 for 2 h, after which the cells were washed three times with 2 mM PBS-EDTA/0.5% bovine serum albumin (BSA), resuspended in RPMI 1640 medium supplemented with 10% FBS, and incubated at 37°C under a 5% CO2 atmosphere. Virus replication was estimated either by measuring the p24 content at different days postinfection with an in-house enzymatic assay (95) (for cells infected with NL4.3Balenv) or by quantifying the percentages of HSA+ cells at 3 dpi (for cells infected with NL4.3Bal-IRES-HSA).
Virus entry assay.
Primary human CD4+ T cells were either left untreated or treated as described above. Next, the cells were inoculated with NL4-3 Balenv (50 ng of p24 per 1 × 105 cells) for 2 h at 37°C. The cells were washed twice with an equal volume of PBS (pH 7.4), resuspended in 100 μl 2.5% trypsin-EDTA (Thermo Fisher Scientific), and incubated for 10 min at 37°C to remove unadsorbed/uninternalized virions. Finally, the cells were washed three times with PBS (pH 7.4) and lysed using 500 μl of disruption buffer (20 mM HEPES [pH 7.4], 150 mM sodium chloride, and 0.5% Triton X-100). Samples were kept at −80°C until they were assayed by the homemade p24 enzymatic test.
Flow cytometry studies.
Surface expression of HSA, CD4, CCR5, CD25, CD69, and CD154 on the total population of purified CD4+ T cells was analyzed by flow cytometry. Briefly, for each experimental condition, 2 × 105 cells were resuspended in 100 μl wash/stain buffer (PBS-2 mM EDTA-0.5% BSA) and stained with the appropriate MAb at optimal concentrations for 15 min at room temperature under dark conditions. The cells were washed twice with wash/stain buffer and resuspended in 2% paraformaldehyde (Thermo Fisher Scientific) prior to flow cytometry analysis (BD FACSCanto; BD Biosciences. Mississauga, ON, Canada). Cellular proliferation assays were performed with the CellTrace CFSE cell proliferation kit (Thermo Fisher Scientific). Briefly, primary human CD4+ T cells were stained with the CFSE dye as specified in the experimental protocol provided with the kit and then stimulated for 72 h with anti-CD3 (clone OKT3; 5 μg/ml) and anti-CD28 (clone 9.3; 2 μg/ml) MAbs in the absence or presence of acetate (20 mM). Fluorescence emission was measured by flow cytometry (BD FACSCanto; BD Biosciences, Mississauga, ON, Canada) to determine the effect of the acetate treatment on cell proliferation.
Analysis of class I/II HDAC activity.
Class I/II HDAC activity was monitored using the HDAC-Glo I/II commercial assay (Promega, Madison, WI). This luminescent assay measures the relative activity of class I/II HDAC from cell extracts. The test uses an acetylated, live-cell-permeant, luminogenic peptide substrate that can be deacetylated by HDAC activities. Deacetylation of the peptide substrate is measured using a coupled enzymatic system in which a protease in the developer reagent cleaves the peptide from aminoluciferin, which is quantified in a reaction using Ultra-Glo recombinant luciferase. In brief, cells were stimulated with anti-CD3 (clone OKT3; 5 μg/ml) and anti-CD28 (clone 9.3; 2 μg/ml) MAbs for 72 h in the absence or presence of acetate (20 mM) or TSA (200 nM) (Promega, Madison, WI), resuspended in PBS-2 mM EDTA (1 × 104 cells/well), and incubated with HDAC-Glow I/II reagent supplemented with 1% Triton X-100 (Sigma-Aldrich, St. Louis, MO) for 30 min at room temperature. Luminescence was measured using a Varioskan Flash multimode reader (Thermo Fisher Scientific). Controls consisted of TZM-bl nuclear extracts because this HeLa-derived cell line contains high endogenous class I/II HDAC activity (unpublished observations).
Quantification of HIV-1 completed reverse transcripts and proviral DNA copies.
Primary human CD4+ T cells (3 × 106 cells/condition) were cultured in RPMI 1640 medium supplemented with 10% FBS at a concentration of 2 × 106 cells/ml, seeded in 48-well flat-bottom tissue culture plates, and treated for 72 h with anti-CD3 (clone OKT3; 5 μg/ml) and anti-CD28 (clone 9.3; 2 μg/ml) MAbs in the absence or presence of acetate (20 mM). NL4.3Balenv virus stocks were treated with DNase I (40 μg/ml; Roche, Bâle, Switzerland) in the presence of MgCl2 (10 mM) and EGTA for 45 min at room temperature prior to their use. Controls consisted of cells treated for 4 h with TSA (200 nM). Next, the cells were infected with HIV-1 for 2 h before being washed three times with PBS-2 mM EDTA-0.5% BSA. The cells were resuspended in RPMI 1640 medium supplemented with 10% FBS and incubated for 24 h to minimize cell-to-cell virus propagation. Genomic DNA was subsequently extracted using a Nucleospin tissue DNA purification kit (Macherey-Nagel, Duren, Germany). Finally, integrated proviral DNA copies were quantified using a combined Alu-HIV-1 PCR and quantitative real-time PCR assay as described previously (85, 96), whereas quantification of completed reverse transcripts was achieved by a quantitative real-time PCR test in a 10-μl reaction mixture containing 20 ng of DNA, TaqMan Fast advanced master mix (Thermo Fisher Scientific), 1 μM (each) of HIV-1-specific sense M667 and antisense M661 primers, and 0.3 μM of the TaqMan probe HIV-5′-carboxyfluorescein (97). Normalization of quantitative real-time PCR data was achieved using the β-globin housekeeping gene. In brief, from every diluted DNA sample, β-globin was quantified using 1 μM of sense (TGGTCTATTTTCCCACCCT) and antisense (TGGCAAAGGTGCCCTTGA) specific primers and 0.3 μM of the TaqMan probe 5′-β-globin-VIC-TCTGTCCACTCCTGATGCTG-NFQ-MGB-3′.
Measurement of histone H3 and H4 acetylation by flow cytometry.
Primary human CD4+ T cells were either left untreated or treated as described above. Next, the cells were washed twice in PBS-2 mM EDTA-0.5% BSA and fixed in 100 μl of 4% formaldehyde for 15 min at room temperature. Controls consisted of cells stimulated with TSA (200 nM) for 4 h before formaldehyde fixation. The cells were washed twice with PBS-2 mM EDTA-0.5% BSA and permeabilized in 100 μl of BD perm/wash buffer for 45 min at 4°C in the dark. The cells were washed twice with PBS-2 mM EDTA-0.5% BSA and stained for 45 min in the dark at room temperature using a mixture of H3 and H4 MAbs. Finally, the cells were washed twice and resuspended in PBS-2 mM EDTA before monitoring the fluorescence intensity by flow cytometry.
Statistical analysis.
Statistical analysis was performed using GraphPad Prism version 6. Ratio-paired Student's t test and one-way analysis of variance (ANOVA) with corrections for multiple comparisons (Dunnett) were performed to define the statistical significance of our results. A threshold P value of ≤0,05 was considered statistically significant, whereas P values of ≤0,01, ≤0,001, and ≤0,0001 were considered highly significant.
ACKNOWLEDGMENTS
We acknowledge the Bioimaging platform of the Infectious Disease Research Centre, which was funded by an equipment and infrastructure grant from the Canadian Foundation for Innovation. We recognize the important contribution of Yann Breton, who provided some virus stocks. The experimental procedure to quantify histone H3 and H4 acetylation was kindly provided by Marion Pardons from the laboratory of Nicolas Chomont (Centre de Recherche du CHUM, Montréal, Canada).
This study was supported by funds allocated to M.J.T. from the Open Operating Grant Program of the Canadian Institutes of Health Research (CIHR) (HOP-143170). M.J.T. is the recipient of the Tier 1 CIHR-Canada Research Chair in Human Immunoretrovirology.
We declare no competing financial interests.
REFERENCES
- 1.Imai K, Inoue H, Tamura M, Cueno ME, Inoue H, Takeichi O, Kusama K, Saito I, Ochiai K. 2012. The periodontal pathogen Porphyromonas gingivalis induces the Epstein-Barr virus lytic switch transactivator ZEBRA by histone modification. Biochimie 94:839–846. doi: 10.1016/j.biochi.2011.12.001. [DOI] [PubMed] [Google Scholar]
- 2.Imai K, Ochiai K. 2011. Role of histone modification on transcriptional regulation and HIV-1 gene expression: possible mechanisms of periodontal diseases in AIDS progression. J Oral Sci 53:1–13. doi: 10.2334/josnusd.53.1. [DOI] [PubMed] [Google Scholar]
- 3.Imai K, Ochiai K, Okamoto T. 2009. Reactivation of latent HIV-1 infection by the periodontopathic bacterium Porphyromonas gingivalis involves histone modification. J Immunol 182:3688–3695. doi: 10.4049/jimmunol.0802906. [DOI] [PubMed] [Google Scholar]
- 4.Imai K, Okamoto T, Ochiai K. 2015. Involvement of Sp1 in butyric acid-induced HIV-1 gene expression. Cell Physiol Biochem 37:853–865. doi: 10.1159/000430213. [DOI] [PubMed] [Google Scholar]
- 5.Imai K, Victoriano AF, Ochiai K, Okamoto T. 2012. Microbial interaction of periodontopathic bacterium Porphyromonas gingivalis and HIV-possible causal link of periodontal diseases to AIDS progression. Curr HIV Res 10:238–244. doi: 10.2174/157016212800618183. [DOI] [PubMed] [Google Scholar]
- 6.Imai K, Yamada K, Tamura M, Ochiai K, Okamoto T. 2012. Reactivation of latent HIV-1 by a wide variety of butyric acid-producing bacteria. Cell Mol Life Sci 69:2583–2592. doi: 10.1007/s00018-012-0936-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Das B, Dobrowolski C, Shahir AM, Feng Z, Yu X, Sha J, Bissada NF, Weinberg A, Karn J, Ye F. 2015. Short chain fatty acids potently induce latent HIV-1 in T-cells by activating P-TEFb and multiple histone modifications. Virology 474:65–81. doi: 10.1016/j.virol.2014.10.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Layden BT, Angueira AR, Brodsky M, Durai V, Lowe WL Jr. 2013. Short chain fatty acids and their receptors: new metabolic targets. Transl Res 161:131–140. doi: 10.1016/j.trsl.2012.10.007. [DOI] [PubMed] [Google Scholar]
- 9.Tan J, McKenzie C, Potamitis M, Thorburn AN, Mackay CR, Macia L. 2014. The role of short-chain fatty acids in health and disease. Adv Immunol 121:91–119. doi: 10.1016/B978-0-12-800100-4.00003-9. [DOI] [PubMed] [Google Scholar]
- 10.Wong JM, de Souza R, Kendall CW, Emam A, Jenkins DJ. 2006. Colonic health: fermentation and short chain fatty acids. J Clin Gastroenterol 40:235–243. doi: 10.1097/00004836-200603000-00015. [DOI] [PubMed] [Google Scholar]
- 11.Nicholson JK, Holmes E, Kinross J, Burcelin R, Gibson G, Jia W, Pettersson S. 2012. Host-gut microbiota metabolic interactions. Science 336:1262–1267. doi: 10.1126/science.1223813. [DOI] [PubMed] [Google Scholar]
- 12.Topping DL, Clifton PM. 2001. Short-chain fatty acids and human colonic function: roles of resistant starch and nonstarch polysaccharides. Physiol Rev 81:1031–1064. [DOI] [PubMed] [Google Scholar]
- 13.Cummings J, Macfarlane G. 1997. Role of intestinal bacteria in nutrient metabolism. J Parenter Enteral Nutr 21:357–365. doi: 10.1177/0148607197021006357. [DOI] [PubMed] [Google Scholar]
- 14.Macfarlane S, Macfarlane GT. 2003. Regulation of short-chain fatty acid production. Proc Nutr Soc 62:67–72. doi: 10.1079/PNS2002207. [DOI] [PubMed] [Google Scholar]
- 15.Cook SI, Sellin JH. 1998. Short chain fatty acids in health and disease. Aliment Pharmacol Ther 12:499–507. doi: 10.1046/j.1365-2036.1998.00337.x. [DOI] [PubMed] [Google Scholar]
- 16.Cronin M, Ventura M, Fitzgerald GF, van Sinderen D. 2011. Progress in genomics, metabolism and biotechnology of bifidobacteria. Int J Food Microbiol 149:4–18. doi: 10.1016/j.ijfoodmicro.2011.01.019. [DOI] [PubMed] [Google Scholar]
- 17.Kim CH, Park J, Kim M. 2014. Gut microbiota-derived short-chain fatty acids, T cells, and inflammation. Immune Netw 14:277–288. doi: 10.4110/in.2014.14.6.277. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Cavaglieri CR, Nishiyama A, Fernandes LC, Curi R, Miles EA, Calder PC. 2003. Differential effects of short-chain fatty acids on proliferation and production of pro- and anti-inflammatory cytokines by cultured lymphocytes. Life Sci 73:1683–1690. doi: 10.1016/S0024-3205(03)00490-9. [DOI] [PubMed] [Google Scholar]
- 19.Wolever TM, Josse RG, Leiter LA, Chiasson JL. 1997. Time of day and glucose tolerance status affect serum short-chain fatty acid concentrations in humans. Metabolism 46:805–811. doi: 10.1016/S0026-0495(97)90127-X. [DOI] [PubMed] [Google Scholar]
- 20.Wolever TM, Schrade KB, Vogt JA, Tsihlias EB, McBurney MI. 2002. Do colonic short-chain fatty acids contribute to the long-term adaptation of blood lipids in subjects with type 2 diabetes consuming a high-fiber diet? Am J Clin Nutr 75:1023–1030. [DOI] [PubMed] [Google Scholar]
- 21.Tarini J, Wolever TM. 2010. The fermentable fibre inulin increases postprandial serum short-chain fatty acids and reduces free-fatty acids and ghrelin in healthy subjects. Appl Physiol Nutr Metab 35:9–16. doi: 10.1139/H09-119. [DOI] [PubMed] [Google Scholar]
- 22.Freeland KR, Wilson C, Wolever TM. 2010. Adaptation of colonic fermentation and glucagon-like peptide-1 secretion with increased wheat fibre intake for 1 year in hyperinsulinaemic human subjects. Br J Nutr 103:82–90. doi: 10.1017/S0007114509991462. [DOI] [PubMed] [Google Scholar]
- 23.Englyst HN, Kingman SM, Cummings JH. 1992. Classification and measurement of nutritionally important starch fractions. Eur J Clin Nutr 46(Suppl 2):S33–S50. [PubMed] [Google Scholar]
- 24.Pan XD, Chen FQ, Wu TX, Tang HG, Zhao ZY. 2009. Prebiotic oligosaccharides change the concentrations of short-chain fatty acids and the microbial population of mouse bowel. J Zhejiang Univ Sci B 10:258–263. doi: 10.1631/jzus.B0820261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Cummings JH, Pomare EW, Branch WJ, Naylor CP, Macfarlane GT. 1987. Short chain fatty acids in human large intestine, portal, hepatic and venous blood. Gut 28:1221–1227. doi: 10.1136/gut.28.10.1221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Bloemen JG, Venema K, van de Poll MC, Olde Damink SW, Buurman WA, Dejong CH. 2009. Short chain fatty acids exchange across the gut and liver in humans measured at surgery. Clin Nutr 28:657–661. doi: 10.1016/j.clnu.2009.05.011. [DOI] [PubMed] [Google Scholar]
- 27.Pomare EW, Branch WJ, Cummings JH. 1985. Carbohydrate fermentation in the human colon and its relation to acetate concentrations in venous blood. J Clin Invest 75:1448–1454. doi: 10.1172/JCI111847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Iwanaga T, Takebe K, Kato I, Karaki S, Kuwahara A. 2006. Cellular expression of monocarboxylate transporters (MCT) in the digestive tract of the mouse, rat, and humans, with special reference to slc5a8. Biomed Res 27:243–254. doi: 10.2220/biomedres.27.243. [DOI] [PubMed] [Google Scholar]
- 29.Tang XL, Wang Y, Li DL, Luo J, Liu MY. 2012. Orphan G protein-coupled receptors (GPCRs): biological functions and potential drug targets. Acta Pharmacol Sin 33:363–371. doi: 10.1038/aps.2011.210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Hirasawa A, Hara T, Katsuma S, Adachi T, Tsujimoto G. 2008. Free fatty acid receptors and drug discovery. Biol Pharm Bull 31:1847–1851. doi: 10.1248/bpb.31.1847. [DOI] [PubMed] [Google Scholar]
- 31.Brown AJ, Goldsworthy SM, Barnes AA, Eilert MM, Tcheang L, Daniels D, Muir AI, Wigglesworth MJ, Kinghorn I, Fraser NJ, Pike NB, Strum JC, Steplewski KM, Murdock PR, Holder JC, Marshall FH, Szekeres PG, Wilson S, Ignar DM, Foord SM, Wise A, Dowell SJ. 2003. The Orphan G protein-coupled receptors GPR41 and GPR43 are activated by propionate and other short chain carboxylic acids. J Biol Chem 278:11312–11319. doi: 10.1074/jbc.M211609200. [DOI] [PubMed] [Google Scholar]
- 32.Le Poul E, Loison C, Struyf S, Springael JY, Lannoy V, Decobecq ME, Brezillon S, Dupriez V, Vassart G, Van Damme J, Parmentier M, Detheux M. 2003. Functional characterization of human receptors for short chain fatty acids and their role in polymorphonuclear cell activation. J Biol Chem 278:25481–25489. doi: 10.1074/jbc.M301403200. [DOI] [PubMed] [Google Scholar]
- 33.Nilsson NE, Kotarsky K, Owman C, Olde B. 2003. Identification of a free fatty acid receptor, FFA2R, expressed on leukocytes and activated by short-chain fatty acids. Biochem Biophys Res Commun 303:1047–1052. doi: 10.1016/S0006-291X(03)00488-1. [DOI] [PubMed] [Google Scholar]
- 34.Halestrap AP, Wilson MC. 2012. The monocarboxylate transporter family—role and regulation. IUBMB Life 64:109–119. doi: 10.1002/iub.572. [DOI] [PubMed] [Google Scholar]
- 35.Cousens LS, Gallwitz D, Alberts BM. 1979. Different accessibilities in chromatin to histone acetylase. J Biol Chem 254:1716–1723. [PubMed] [Google Scholar]
- 36.Davie JR. 2003. Inhibition of histone deacetylase activity by butyrate. J Nutr 133:2485S–2493S. [DOI] [PubMed] [Google Scholar]
- 37.Singh N, Thangaraju M, Prasad PD, Martin PM, Lambert NA, Boettger T, Offermanns S, Ganapathy V. 2010. Blockade of dendritic cell development by bacterial fermentation products butyrate and propionate through a transporter (Slc5a8)-dependent inhibition of histone deacetylases. J Biol Chem 285:27601–27608. doi: 10.1074/jbc.M110.102947. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Meijer K, de Vos P, Priebe MG. 2010. Butyrate and other short-chain fatty acids as modulators of immunity: what relevance for health? Curr Opin Clin Nutr Metab Care 13:715–721. doi: 10.1097/MCO.0b013e32833eebe5. [DOI] [PubMed] [Google Scholar]
- 39.Biel M, Wascholowski V, Giannis A. 2005. Epigenetics—an epicenter of gene regulation: histones and histone-modifying enzymes. Angew Chem Int Ed Engl 44:3186–3216. doi: 10.1002/anie.200461346. [DOI] [PubMed] [Google Scholar]
- 40.Waldecker M, Kautenburger T, Daumann H, Busch C, Schrenk D. 2008. Inhibition of histone-deacetylase activity by short-chain fatty acids and some polyphenol metabolites formed in the colon. J Nutr Biochem 19:587–593. doi: 10.1016/j.jnutbio.2007.08.002. [DOI] [PubMed] [Google Scholar]
- 41.Sheridan PL, Mayall TP, Verdin E, Jones KA. 1997. Histone acetyltransferases regulate HIV-1 enhancer activity in vitro. Genes Dev 11:3327–3340. doi: 10.1101/gad.11.24.3327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Imai K, Okamoto T. 2006. Transcriptional repression of human immunodeficiency virus type 1 by AP-4. J Biol Chem 281:12495–12505. doi: 10.1074/jbc.M511773200. [DOI] [PubMed] [Google Scholar]
- 43.Jiang G, Espeseth A, Hazuda DJ, Margolis DM. 2007. c-Myc and Sp1 contribute to proviral latency by recruiting histone deacetylase 1 to the human immunodeficiency virus type 1 promoter. J Virol 81:10914–10923. doi: 10.1128/JVI.01208-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Williams SA, Chen LF, Kwon H, Ruiz-Jarabo CM, Verdin E, Greene WC. 2006. NF-kappaB p50 promotes HIV latency through HDAC recruitment and repression of transcriptional initiation. EMBO J 25:139–149. doi: 10.1038/sj.emboj.7600900. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Coull JJ, Romerio F, Sun JM, Volker JL, Galvin KM, Davie JR, Shi Y, Hansen U, Margolis DM. 2000. The human factors YY1 and LSF repress the human immunodeficiency virus type 1 long terminal repeat via recruitment of histone deacetylase 1. J Virol 74:6790–6799. doi: 10.1128/JVI.74.15.6790-6799.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Van Lint C, Emiliani S, Ott M, Verdin E. 1996. Transcriptional activation and chromatin remodeling of the HIV-1 promoter in response to histone acetylation. EMBO J 15:1112–1120. [PMC free article] [PubMed] [Google Scholar]
- 47.Kashanchi F, Melpolder JC, Epstein JS, Sadaie MR. 1997. Rapid and sensitive detection of cell-associated HIV-1 in latently infected cell lines and in patient cells using sodium-n-butyrate induction and RT-PCR. J Med Virol 52:179–189. [PubMed] [Google Scholar]
- 48.Wilson MJ, Hall V, Brazier J, Lewis MA. 2000. Evaluation of a phenotypic scheme for identification of the ‘butyrate-producing’ Peptostreptococcus species. J Med Microbiol 49:747–751. doi: 10.1099/0022-1317-49-8-747. [DOI] [PubMed] [Google Scholar]
- 49.Kantor B, Ma H, Webster-Cyriaque J, Monahan PE, Kafri T. 2009. Epigenetic activation of unintegrated HIV-1 genomes by gut-associated short chain fatty acids and its implications for HIV infection. Proc Natl Acad Sci U S A 106:18786–18791. doi: 10.1073/pnas.0905859106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Wagner TA, McKernan JL, Tobin NH, Tapia KA, Mullins JI, Frenkel LM. 2013. An increasing proportion of monotypic HIV-1 DNA sequences during antiretroviral treatment suggests proliferation of HIV-infected cells. J Virol 87:1770–1778. doi: 10.1128/JVI.01985-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Wagner TA, McLaughlin S, Garg K, Cheung CY, Larsen BB, Styrchak S, Huang HC, Edlefsen PT, Mullins JI, Frenkel LM. 2014. HIV latency. Proliferation of cells with HIV integrated into cancer genes contributes to persistent infection. Science 345:570–573. doi: 10.1126/science.1256304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Ribeiro RM, Mohri H, Ho DD, Perelson AS. 2002. In vivo dynamics of T cell activation, proliferation, and death in HIV-1 infection: why are CD4+ but not CD8+ T cells depleted? Proc Natl Acad Sci U S A 99:15572–15577. doi: 10.1073/pnas.242358099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Cleret-Buhot A, Zhang Y, Planas D, Goulet JP, Monteiro P, Gosselin A, Wacleche VS, Tremblay CL, Jenabian MA, Routy JP, El-Far M, Chomont N, Haddad EK, Sekaly RP, Ancuta P. 2015. Identification of novel HIV-1 dependency factors in primary CCR4CCR6Th17 cells via a genome-wide transcriptional approach. Retrovirology 12:102. doi: 10.1186/s12977-015-0226-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Aoyama M, Kotani J, Usami M. 2010. Butyrate and propionate induced activated or non-activated neutrophil apoptosis via HDAC inhibitor activity but without activating GPR-41/GPR-43 pathways. Nutrition 26:653–661. doi: 10.1016/j.nut.2009.07.006. [DOI] [PubMed] [Google Scholar]
- 55.Hinnebusch BF, Meng S, Wu JT, Archer SY, Hodin RA. 2002. The effects of short-chain fatty acids on human colon cancer cell phenotype are associated with histone hyperacetylation. J Nutr 132:1012–1017. [DOI] [PubMed] [Google Scholar]
- 56.Golub EI, Li GR, Volsky DJ. 1991. Induction of dormant HIV-1 by sodium butyrate: involvement of the TATA box in the activation of the HIV-1 promoter. AIDS 5:663–668. doi: 10.1097/00002030-199106000-00004. [DOI] [PubMed] [Google Scholar]
- 57.Agosto LM, Gagne M, Henderson AJ. 2015. Impact of chromatin on HIV replication. Genes 6:957–976. doi: 10.3390/genes6040957. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Goncalves J, Moreira E, Sequeira IJ, Rodrigues AS, Rueff J, Bras A. 2016. Integration of HIV in the human genome: which sites are preferential? A genetic and statistical assessment. Int J Genomics 2016:2168590. doi: 10.1155/2016/2168590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Dillon SM, Lee EJ, Donovan AM, Guo K, Harper MS, Frank DN, McCarter MD, Santiago ML, Wilson CC. 2016. Enhancement of HIV-1 infection and intestinal CD4+ T cell depletion ex vivo by gut microbes altered during chronic HIV-1 infection. Retrovirology 13:5. doi: 10.1186/s12977-016-0237-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Zevin AS, McKinnon L, Burgener A, Klatt NR. 2016. Microbial translocation and microbiome dysbiosis in HIV-associated immune activation. Curr Opin HIV AIDS 11:182–190. doi: 10.1097/COH.0000000000000234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Dinh DM, Volpe GE, Duffalo C, Bhalchandra S, Tai AK, Kane AV, Wanke CA, Ward HD. 2015. Intestinal microbiota, microbial translocation, and systemic inflammation in chronic HIV infection. J Infect Dis 211:19–27. doi: 10.1093/infdis/jiu409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Klatt NR, Funderburg NT, Brenchley JM. 2013. Microbial translocation, immune activation, and HIV disease. Trends Microbiol 21:6–13. doi: 10.1016/j.tim.2012.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Brenchley JM, Douek DC. 2012. Microbial translocation across the GI tract. Annu Rev Immunol 30:149–173. doi: 10.1146/annurev-immunol-020711-075001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Marchetti G, Cozzi-Lepri A, Merlini E, Bellistri GM, Castagna A, Galli M, Verucchi G, Antinori A, Costantini A, Giacometti A, di Caro A, D'Arminio Monforte A, ICONA Foundation Study Group . 2011. Microbial translocation predicts disease progression of HIV-infected antiretroviral-naive patients with high CD4+ cell count. AIDS 25:1385–1394. doi: 10.1097/QAD.0b013e3283471d10. [DOI] [PubMed] [Google Scholar]
- 65.Cummings JH. 1981. Short chain fatty acids in the human colon. Gut 22:763–779. doi: 10.1136/gut.22.9.763. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Hijova E, Chmelarova A. 2007. Short chain fatty acids and colonic health. Bratisl Lek Listy 108:354–358. [PubMed] [Google Scholar]
- 67.Binder HJ. 2010. Role of colonic short-chain fatty acid transport in diarrhea. Annu Rev Physiol 72:297–313. doi: 10.1146/annurev-physiol-021909-135817. [DOI] [PubMed] [Google Scholar]
- 68.Mattapallil JJ, Douek DC, Hill B, Nishimura Y, Martin M, Roederer M. 2005. Massive infection and loss of memory CD4+ T cells in multiple tissues during acute SIV infection. Nature 434:1093–1097. doi: 10.1038/nature03501. [DOI] [PubMed] [Google Scholar]
- 69.Stevenson M, Stanwick TL, Dempsey MP, Lamonica CA. 1990. HIV-1 replication is controlled at the level of T cell activation and proviral integration. EMBO J 9:1551–1560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Szeto GL, Brice AK, Yang HC, Barber SA, Siliciano RF, Clements JE. 2010. Minocycline attenuates HIV infection and reactivation by suppressing cellular activation in human CD4+ T cells. J Infect Dis 201:1132–1140. doi: 10.1086/651277. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Levring TB, Hansen AK, Nielsen BL, Kongsbak M, von Essen MR, Woetmann A, Odum N, Bonefeld CM, Geisler C. 2012. Activated human CD4+ T cells express transporters for both cysteine and cystine. Sci Rep 2:266. doi: 10.1038/srep00266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Freedman BD, Price MA, Deutsch CJ. 1992. Evidence for voltage modulation of IL-2 production in mitogen-stimulated human peripheral blood lymphocytes. J Immunol 149:3784–3794. [PubMed] [Google Scholar]
- 73.Koo GC, Blake JT, Talento A, Nguyen M, Lin S, Sirotina A, Shah K, Mulvany K, Hora D Jr, Cunningham P, Wunderler DL, McManus OB, Slaughter R, Bugianesi R, Felix J, Garcia M, Williamson J, Kaczorowski G, Sigal NH, Springer MS, Feeney W. 1997. Blockade of the voltage-gated potassium channel Kv1.3 inhibits immune responses in vivo. J Immunol 158:5120–5128. [PubMed] [Google Scholar]
- 74.Beeton C, Wulff H, Barbaria J, Clot-Faybesse O, Pennington M, Bernard D, Cahalan MD, Chandy KG, Beraud E. 2001. Selective blockade of T lymphocyte K(+) channels ameliorates experimental autoimmune encephalomyelitis, a model for multiple sclerosis. Proc Natl Acad Sci U S A 98:13942–13947. doi: 10.1073/pnas.241497298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Urrego D, Tomczak AP, Zahed F, Stuhmer W, Pardo LA. 2014. Potassium channels in cell cycle and cell proliferation. Philos Trans R Soc Lond B Biol Sci 369:20130094. doi: 10.1098/rstb.2013.0094. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Roy J, Denovan-Wright EM, Linsdell P, Cowley EA. 2006. Exposure to sodium butyrate leads to functional downregulation of calcium-activated potassium channels in human airway epithelial cells. Pflugers Arch 453:167–176. doi: 10.1007/s00424-006-0128-8. [DOI] [PubMed] [Google Scholar]
- 77.Clapham PR, McKnight A. 2001. HIV-1 receptors and cell tropism. Br Med Bull 58:43–59. doi: 10.1093/bmb/58.1.43. [DOI] [PubMed] [Google Scholar]
- 78.Matalon S, Palmer BE, Nold MF, Furlan A, Kassu A, Fossati G, Mascagni P, Dinarello CA. 2010. The histone deacetylase inhibitor ITF2357 decreases surface CXCR4 and CCR5 expression on CD4(+) T-cells and monocytes and is superior to valproic acid for latent HIV-1 expression in vitro. J Acquir Immune Defic Syndr 54:1–9. doi: 10.1097/QAI.0b013e3181d3dca3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Bleul CC, Wu L, Hoxie JA, Springer TA, Mackay CR. 1997. The HIV coreceptors CXCR4 and CCR5 are differentially expressed and regulated on human T lymphocytes. Proc Natl Acad Sci U S A 94:1925–1930. doi: 10.1073/pnas.94.5.1925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Dragic T, Litwin V, Allaway GP, Martin SR, Huang Y, Nagashima KA, Cayanan C, Maddon PJ, Koup RA, Moore JP, Paxton WA. 1996. HIV-1 entry into CD4+ cells is mediated by the chemokine receptor CC-CKR-5. Nature 381:667–673. doi: 10.1038/381667a0. [DOI] [PubMed] [Google Scholar]
- 81.Deng H, Liu R, Ellmeier W, Choe S, Unutmaz D, Burkhart M, Di Marzio P, Marmon S, Sutton RE, Hill CM, Davis CB, Peiper SC, Schall TJ, Littman DR, Landau NR. 1996. Identification of a major co-receptor for primary isolates of HIV-1. Nature 381:661–666. doi: 10.1038/381661a0. [DOI] [PubMed] [Google Scholar]
- 82.Schroder AR, Shinn P, Chen H, Berry C, Ecker JR, Bushman F. 2002. HIV-1 integration in the human genome favors active genes and local hotspots. Cell 110:521–529. doi: 10.1016/S0092-8674(02)00864-4. [DOI] [PubMed] [Google Scholar]
- 83.Rezaei SD, Cameron PU. 2015. Human immunodeficiency virus (HIV)-1 integration sites in viral latency. Curr HIV/AIDS Rep 12:88–96. doi: 10.1007/s11904-014-0241-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Han Y, Lassen K, Monie D, Sedaghat AR, Shimoji S, Liu X, Pierson TC, Margolick JB, Siliciano RF, Siliciano JD. 2004. Resting CD4+ T cells from human immunodeficiency virus type 1 (HIV-1)-infected individuals carry integrated HIV-1 genomes within actively transcribed host genes. J Virol 78:6122–6133. doi: 10.1128/JVI.78.12.6122-6133.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Suzuki Y, Misawa N, Sato C, Ebina H, Masuda T, Yamamoto N, Koyanagi Y. 2003. Quantitative analysis of human immunodeficiency virus type 1 DNA dynamics by real-time PCR: integration efficiency in stimulated and unstimulated peripheral blood mononuclear cells. Virus Genes 27:177–188. doi: 10.1023/A:1025732728195. [DOI] [PubMed] [Google Scholar]
- 86.Gorisch SM, Wachsmuth M, Toth KF, Lichter P, Rippe K. 2005. Histone acetylation increases chromatin accessibility. J Cell Sci 118:5825–5834. doi: 10.1242/jcs.02689. [DOI] [PubMed] [Google Scholar]
- 87.Craigie R, Bushman FD. 2012. HIV DNA integration. Cold Spring Harb Perspect Med 2:a006890. doi: 10.1101/cshperspect.a006890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Platt EJ, Bilska M, Kozak SL, Kabat D, Montefiori DC. 2009. Evidence that ecotropic murine leukemia virus contamination in TZM-bl cells does not affect the outcome of neutralizing antibody assays with human immunodeficiency virus type 1. J Virol 83:8289–8292. doi: 10.1128/JVI.00709-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Wei X, Decker JM, Liu H, Zhang Z, Arani RB, Kilby JM, Saag MS, Wu X, Shaw GM, Kappes JC. 2002. Emergence of resistant human immunodeficiency virus type 1 in patients receiving fusion inhibitor (T-20) monotherapy. Antimicrob Agents Chemother 46:1896–1905. doi: 10.1128/AAC.46.6.1896-1905.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Derdeyn CA, Decker JM, Sfakianos JN, Wu X, O'Brien WA, Ratner L, Kappes JC, Shaw GM, Hunter E. 2000. Sensitivity of human immunodeficiency virus type 1 to the fusion inhibitor T-20 is modulated by coreceptor specificity defined by the V3 loop of gp120. J Virol 74:8358–8367. doi: 10.1128/JVI.74.18.8358-8367.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Platt EJ, Wehrly K, Kuhmann SE, Chesebro B, Kabat D. 1998. Effects of CCR5 and CD4 cell surface concentrations on infections by macrophagetropic isolates of human immunodeficiency virus type 1. J Virol 72:2855–2864. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Takeuchi Y, McClure MO, Pizzato M. 2008. Identification of gammaretroviruses constitutively released from cell lines used for human immunodeficiency virus research. J Virol 82:12585–12588. doi: 10.1128/JVI.01726-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Dornadula G, Zhang H, Shetty S, Pomerantz RJ. 1999. HIV-1 virions produced from replicating peripheral blood lymphocytes are more infectious than those from nonproliferating macrophages due to higher levels of intravirion reverse transcripts: implications for pathogenesis and transmission. Virology 253:10–16. doi: 10.1006/viro.1998.9465. [DOI] [PubMed] [Google Scholar]
- 94.Imbeault M, Lodge R, Ouellet M, Tremblay MJ. 2009. Efficient magnetic bead-based separation of HIV-1-infected cells using an improved reporter virus system reveals that p53 up-regulation occurs exclusively in the virus-expressing cell population. Virology 393:160–167. doi: 10.1016/j.virol.2009.07.009. [DOI] [PubMed] [Google Scholar]
- 95.Bounou S, Leclerc JE, Tremblay MJ. 2002. Presence of host ICAM-1 in laboratory and clinical strains of human immunodeficiency virus type 1 increases virus infectivity and CD4(+)-T-cell depletion in human lymphoid tissue, a major site of replication in vivo. J Virol 76:1004–1014. doi: 10.1128/JVI.76.3.1004-1014.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Thibault S, Fromentin R, Tardif MR, Tremblay MJ. 2009. TLR2 and TLR4 triggering exerts contrasting effects with regard to HIV-1 infection of human dendritic cells and subsequent virus transfer to CD4+ T cells. Retrovirology 6:42. doi: 10.1186/1742-4690-6-42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Zack JA, Arrigo SJ, Weitsman SR, Go AS, Haislip A, Chen IS. 1990. HIV-1 entry into quiescent primary lymphocytes: molecular analysis reveals a labile, latent viral structure. Cell 61:213–222. doi: 10.1016/0092-8674(90)90802-L. [DOI] [PubMed] [Google Scholar]






