Abstract
HIV invades the brain early after infection; however, its interactions with the cells of the blood–brain barrier (BBB) remain poorly understood. Our goal was to evaluate the role of occludin, one of the tight junction proteins that regulate BBB functions in HIV infection of BBB pericytes. We provide evidence that occludin levels largely control the metabolic responses of human pericytes to HIV. Occludin in BBB pericytes decreased by 10% during the first 48 h after HIV infection, correlating with increased nuclear translocation of the gene repressor C-terminal-binding protein (CtBP)-1 and NFκB-p65 activation. These changes were associated with decreased expression and activation of the class III histone deacetylase sirtuin (SIRT)-1. Occludin levels recovered 96 h after infection, restoring SIRT-1 and reducing HIV transcription to 20% of its highest values. We characterized occludin biochemically as a novel NADH oxidase that controls the expression and activation of SIRT-1. The inverse correlation between occludin and HIV transcription was then replicated in human primary macrophages and differentiated monocytic U937 cells, in which occludin silencing resulted in 75 and 250% increased viral transcription, respectively. Our work shows that occludin has previously unsuspected metabolic properties and is a target of HIV infection, opening the possibility of designing novel pharmacological approaches to control HIV transcription.—Castro, V., Bertrand, L., Luethen, M., Dabrowski, S., Lombardi, J., Morgan, L., Sharova, N., Stevenson, M., Blasig, I. E., Toborek, M. Occludin controls HIV transcription in brain pericytes via regulation of SIRT-1 activation.
Keywords: blood–brain barrier, human macrophage, metabolic regulation, NADH oxidase, tight junction protein
HIV enters the brain shortly after infection, causing disruption of the blood–brain barrier (BBB) characterized by structural and functional impairment of tight junctions (TJs) and cell invasion of endothelial cells, astrocytes, and pericytes. To date, little is known about the impact of HIV on pericytes. These cells form a functionally heterogeneous population that expresses specific molecular markers (1, 2), but also shares markers with vascular smooth muscle (3) and endothelial cells (4). Pericytes share functional immunologic similarities with microglia and perivascular macrophages, but express a different set of inflammatory molecules (5). We recently showed that BBB pericytes express the chemokine receptors CXCR4 and CCR5, enabling their infection by cell-free HIV-1 (6); however, their molecular response to HIV remains poorly understood.
Once HIV enters the cell, the proviral genome is translocated to the nucleus and integrated into the host cell genome to be regulated by cellular and viral transcription factors. The viral core promoter contains 2 binding sites for NF-κB, which plays a central role in the proviral activation pathway and stimulation of transcriptional elongation by the p65 subunit (7). The activity of NFκB is modulated by acetylation, and reduction in acetylated-NFκB correlates with lower HIV transcription (8). NFκB is deacetylated by the NAD+-dependent class III histone deacetylase SIRT-1 (9), whose activation is markedly decreased during HIV infection (10). In endothelial cells, HIV avidly represses transcription and promotes rapid degradation of occludin (11), a protein also expressed in pericytes (4).
Occludin is canonically considered a TJ molecule and marker of epithelial and endothelial cells, where its phosphorylation triggers changes in TJ structure and function (12); furthermore, altered occludin expression has been linked to dedifferentiation and cancer progression and invasiveness. However, the molecular mechanisms underlying these functional implications remain elusive. Occludin C-terminal domain is relevant for those functions, as it ends in a 96 aa-long coiled–coil (CC) domain that binds the scaffolding proteins zonula occludens (ZO)-1, ZO-2, ZO-3, F-actin, VAP33, and cingulin, as well as regulatory proteins, such as tyrosine kinase c-Yes, the p58 subunit of PI3K, PKC, and connexin-26 (12). By associating with ZO-1, occludin is indirectly linked to other TJ molecules (claudins and junctional adhesion molecules), G proteins, the transcription factor ZONAB, and the actin cytoskeleton (13).
The targeting of occludin by HIV and its relationship to signaling and structural molecules, prompt the possibility that occludin plays a physiologic role in combatting HIV. In this study, we address the relationship between HIV infection and occludin expression in BBB pericytes and present evidence characterizing occludin as a novel NADH oxidase that controls SIRT-1 expression and activation, inhibiting HIV transcription.
MATERIALS AND METHODS
Cell cultures
Primary human brain capillary pericytes (ScienCell, Carlsbad, CA, USA) belonging to different batches and donors were cultured with 5% CO2 at 37°C in serum- and growth factor–complemented growth medium (ScienCell) and used from passages 2 to 7. Pericyte cultures were characterized as positive for αSMA, NG2, HLA-DRIII, and cluster of differentiation (CD)13, -14, -34, -45, -73, -79, and -146, but negative for desmin and CD90 and -105. Human embryonic kidney (HEK)-293 cells were cultured up to 20 passages. U937 cells were differentiated with 20 nM phorbol-12-myristate-13-acetate for 24 h and cultured with RPMI enriched with 10% fetal bovine serum (Thermo Scientific–Life Technologies, Carlsbad, CA, USA). Human peripheral blood monocytes obtained from different donors were cultured at 37°C for 3 d in DMEM enriched with 10% pooled true human serum, 1% l-glutamine, 0.1% gentamicin, and 6 ng/ml macrophage colony-stimulating factor (M-CSF) to induce differentiation. Medium was later exchanged for a similar growth medium but without M-CSF, and cells were incubated for an additional 4 d before use in experiments.
HIV production, infection, and quantitation
HIV1-NL43 proviral DNA, also encoding interdomain green fluorescent protein (iGFP), was produced by transfection of HEK-293T/17 cells. The infection procedure was performed with cell-free virus (6). Transcription rates were quantified by measuring GFP fluorescence with excitation set at 390 nm, cutoff at 495 nm, and spectral detection in the 500–570 nm range. Representative spectra are shown in Supplemental Fig. S2.
Protein quantitation by spectrally resolved in-cell ELISA
Cells were cultured in 96-well plates, stained with DNA/RNA-binding anthraquinone (DRAQ)-5 (1:50; Cell Signaling Technology, Danvers, MA, USA), and fixed with ice-cold methanol. HIV-infected cells were fixed with ice-cold methanol and acetone (50:50 v/v). Nonspecific binding sites were blocked with 5% fat-free milk in Tris-buffered saline–Tween 20. The cells were immunolabeled for 2 h at room temperature with primary antibodies and then incubated with fluorescently labeled secondary antibodies. The following primary antibodies and dilutions (in PBS) were used: total and phospho-Ser47 SIRT-1, mouse anti-NFκB-p65, and rabbit anti-NFκB-p65 AcK310 (all 1:100; Abcam, Cambridge, MA, USA) and mouse monoclonal anti-occludin (1:50; clone OC-3F10, Alexa-Fluor-594 conjugated; Thermo Scientific–Life Technologies). The following secondary antibodies were used at a 1:100 dilution in PBS: donkey anti-goat, anti-mouse (Dylight-680 and -800; Thermo Scientific–Pierce, Rockford, IL, USA) and anti-rabbit (Dylight-680, Dylight-800, or Alexa Fluor 405 and Pacific Blue) or goat anti-mouse (Alexa-Fluor-488; both from Thermo Scientific–Life Technologies). Readings were performed to detect and quantify the fluorescence spectral intensity of the bound antibodies. The spectrally resolved in-cell ELISA (SPRICE) data were normalized to DRAQ5 fluorescence. Representative spectra are shown in Supplemental Fig. S2.
Detection of NADH in living cells
Live-cell fluorescence spectroscopy was performed in pericytes. NADH was quantified by excitation at 350 nm, and its emission spectrum was recorded from 430 to 470 nm. Living HEK-293 cells were analyzed by confocal microscopy (510 LSM META equipped with nonlinear optics; Zeiss, Jena, Germany). NADH was excited with a titanium-sapphire biphotonic infrared laser locked at 720 nm, and its fluorescence was detected spectrally at 436–475 nm. Representative spectra are shown in Supplemental Fig. S3. Images were quantified in ImageJ [National Institutes of Health (NIH), Bethesda, MD, USA].
Quantitation of NADH oxidase activity
Treated cultures were scrapped on ice in extraction buffer containing 1% Triton X-100, 25 mM 2-[4-(2-hydroxyethyl)piperazin-1-yl]ethanesulfonic acid (HEPES)/NaOH (pH 7.4), 150 mM NaCl, 4 mM EDTA, and 1:500 protease inhibitor cocktail; transferred into Eppendorf (Hamburg, Germany) tubes; sonicated; and centrifuged. Protein concentration was determined by bicinchoninic acid (BCA) assay (Thermo Scientific-Pierce). Supernatants were diluted with aqueous solutions of NADH to achieve a final concentration of 3.5 mg/ml protein and a 5–500 µM range of NADH. Light absorbance at 340 and 260 nm was used to measure NADH and NAD+, respectively.
Occludin purification and determination of enzymatic activity
The CC domain of murine occludin (aa 264–521) was generated recombinantly in Escherichia coli and purified (14). The purified full C-terminal domain of human occludin was obtained commercially (TrueORFGold RC206468, codons 266–522; OriGene, Rockville, MD, USA). SDS-PAGE images of both constructs are shown in Supplemental Fig. S3.
To measure enzymatic activity, we diluted occludin fragments in elution buffer and added to them to a series of NADH aqueous solutions. Spectral absorbance was acquired for NAD+. Kinetic parameters were obtained by diluting the occludin/NADH solutions in reaction buffer and detecting NAD+ light absorbance every 30 s for 15 min.
Occludin depletion and overexpression
Occludin was silenced by transfection (Lipofectamine 2000; Thermo Scientific-Life Technologies) with 6.25 pM per well anti-occludin 27-mer human small interfering (si)RNA (Trilencer-2, UGCACCAAGCAAUGACAUAUAUGGT; OriGene) in RNA resuspension buffer (100 mM KAc and 30 mM HEPES; pH 7.0). Control siRNA [Trilencer-27 universal scrambled (SCR) negative control siRNA; OriGene] or custom made control (AAAGAGCGACUUUACSCACdTdT; Dharmacon, Lafayette, CO, USA) was used. Typical siRNA incubation lasted 16 h, as occludin’s half-life is 11 h.
For occludin overexpression experiments, cells grown to 75% confluence were transfected (Lipofectamine 2000) with N-terminally tagged occludin with yellow fluorescent protein (YFP) or C-terminally tagged occludin with DDK (TrueORFGold RC206468; Origene). Occludin levels were quantified by SPRICE. Representative spectral recordings and immunoblots of occludin are shown in Supplemental Figs. S2 and S3, respectively.
Immunofluorescence, confocal microscopy, and immunoblot analysis
Cells were fixed and processed for immunofluorescence (6). Occludin was labeled with an Alexa Fluor 594–conjugated monoclonal anti-occludin (C-terminal domain) antibody (1:100; Thermo–Scientific–Life Technologies). C-terminal-binding protein (CtBP)-1 was detected with an anti-CtBP1 antibody (1:200; Abcam) paired with a secondary anti-rabbit antibody conjugated with Pacific Blue (Thermo Scientific–Life Technologies). Cell nuclei were stained with DRAQ5 (1:200; Abcam). Live-cell imaging was performed on HEK-293 cells, with the cell membranes stained with 0.5% trypan blue, on an LSM 510 confocal microscope (Zeiss) equipped with a spectral detector. HEK-293 cells expressing YFP-tagged occludin were used as the control.
For immunoblot analysis, the cells were lysed, diluted in denaturing protein loading dye, heated, and loaded onto 5–20% SDS-Tris glycine extended (TGX) precast gels (Bio-Rad, Hercules, CA, USA) (15). Samples were transferred to PVDF membranes, immunolabeled, and detected by chemiluminescence. Representative immunoblots of NFκB-p65 and SIRT-1 are shown in Supplemental Fig. S2.
In silico analysis
Predictions of the tridimensional structure of the C-terminal domain of occludin (fragments aa 266–522, 266–415, and 323–522) were performed with I-TASSER (http://zhanglab.ccmb.med.umich.edu/I-TASSER/; University of Michigan, Ann Arbor, MI, USA). Prediction of the putative binding partners of the same fragments was performed with the protein ligand–binding site prediction server COACH (http://zhanglab.ccmb.med.umich.edu/COACH/; University of Michigan). Sequence similarity searches and homology were performed with BLAST (http://blast.ncbi.nlm.nih.gov/Blast.cgi; National Center for Biotechnology Information, NIH). Docking of NADH in occludin was calculated with AutoDock Vina (Scripps Research Institute, San Diego, CA, USA). UCSF Chimera (UCSF Resource for Biocomputing, University of California, San Francisco, CA, USA) was used for molecular visualization.
Statistical analysis
Data were tested for normality with the D’Agostino-Pearson and Shapiro-Wilk tests with a confidence interval of 99%. For univariate datasets, a 1-way ANOVA, and for nonparametric data, the Kruskal-Wallis test, were performed to determine the probabilities. Groups to be compared were selected during experimental design, and the Fisher least-squares difference post hoc test was used to compare such groups. Multivariate datasets were analyzed with 2-way ANOVA. Binary datasets were compared by using the Welch-corrected t test or Mann-Whitney U test if the data were nonparametric. Significance was considered at P = 0.01 for all cases.
RESULTS
HIV replication inversely correlates with occludin levels in human BBB pericytes
Brain pericytes were infected with cell-free HIV-1, engineered to encode GFP as a transcriptional reporter. The maximum transcription rate occurred at 48 h, decreased thereafter to a minimum rate reached at 96 h, and remained stable after that time point (Fig. 1A). Viral replication rates measured by HIV-specific RT assay were consistent with the observed decrease in HIV replication rate (Fig. 1B). Levels of transcriptionally active (acetylated) NFκB-p65 peaked at 48 h after infection and decreased thereafter, correlating with the observed changes in HIV transcriptional efficiency (Fig. 1C). We also investigated the dynamics of occludin expression in HIV-infected pericytes. Occludin was depleted 48 h after infection, recovered at 96 h, and was overexpressed at 120 h (Fig. 1D, 1E), inversely mirroring the changes in HIV replication rates. Using specific protein kinase inhibitors, we found that the activity of p38-MAPK was needed for the initial elevation in HIV transcription and concurrent loss of occludin 48 h after infection, whereas ROCK activity was active in the subsequent occludin recovery and decreased HIV transcription (Supplemental Fig. S1A–D). Autocrine modulation of HIV transcription by interferons was excluded, as IFN-α, -β, and -γ levels remained unaffected at 48 h after infection (Supplemental Fig. S1E).
Figure 1.
HIV-1 replication in BBB pericytes is biphasic and correlates inversely with occludin expression levels. A) Quantitation of HIV transcription rate by quantitation of GFP encoded in the viral genome at the indicated postinfection times. Data are means ± sem. B) Decrease in viral replication rates was supported by HIV-1-specific RT activity assay at the indicated times. C) Levels of acetylated NFκB-p65 in pericytes at the indicated postinfection times, as measured by SPRICE. D) SPRICE quantitation of occludin in HIV-infected pericytes at the indicated times. The levels of occludin in infected cells were compared to the occludin content in noninfected pericytes cultured in parallel. E) Confocal microscopy of occludin immunolocalized in HIV-infected pericytes 120 after infection. NI, noninfected. F) Representative confocal microscopy images of occludin immunolocalized in pericytes treated with siRNA against occludin (OCC−) or SCR-treated or WT cells, 12 h after silencing. Data (AU) indicate the average fluorescence intensity of immunolabeled occludin in the respective image. G) Representative confocal microscopy images of HIV-infected but otherwise similarly treated pericytes 48 h after infection, performed 12 h after occludin silencing. Nuclei were stained with DRAQ5 (red). GFP fluorescence (green) reflects the rate of HIV transcription. Data (AU) indicate GFP fluorescence intensities quantified in the respective images. H) Confocal microscopy images of WT-pericytes exposed for 48 h to the supernatants of the HIV-infected WT or OCC− pericytes shown in (G). I) Regression analysis of the relative change in transcriptional efficiency (red) measured by GFP quantitation and amount of virus released into the supernatants (blue), measured by RT assay 48 h after WT pericytes were infected with HIV at various viral loads (n = 3). J) Representative confocal microscopy images of WT pericytes 48 h after exposure to the indicated HIV inocula, showing GFP fluorescence. AU, arbitrary units.
Occludin depletion causes a robust increase in viral transcription
To explore the possibility that it negatively influences HIV transcription, occludin was silenced, followed by HIV infection and evaluation of its transcription efficiency by GFP quantitation. Reduction of occludin levels induced an increase in HIV transcriptional efficiency 48 h after infection when compared to similarly infected wild-type (WT) and SCR negative control siRNA–treated pericytes (Fig. 1F, G). Culturing WT pericytes with the supernatants of HIV-infected/occludin-deficient pericytes resulted in a 3-fold increase in viral transcription, as compared to WT pericytes cultured with supernatants of HIV-infected/WT pericytes (Fig. 1H). Thus, relatively low variations in viral inoculum can result in much larger changes in the transcriptional rates of infected cells.
This possibility was further explored by infecting WT pericytes with different loads of virus originating from a single stock. Increasing the viral inoculum over a 200% range [100,000–350,000 scpm (scintillation counts per minute)] resulted in and elevated viral release into the supernatants by only 27% at 48 h after infection, whereas transcriptional efficiencies increased >200% (Fig. 1I). At lower inoculation titers (100,000–200,000 scpm) the rate of increase in viral release was more accentuated than that at higher titers, whereas elevated transcriptional efficiency was prominent only at higher inoculation titers when the rate of viral release stabilized (Fig. 1J). These findings indicate that relatively small differences in HIV release from occludin-depleted pericytes can cause a robust increase in viral transcription in newly infected cells.
Occludin levels regulate SIRT-1 expression and activation
Because HIV transcriptional efficiency is associated with NFκB activity (16), which, in turn, is modulated by the histone deacetylase SIRT-1 (17), the impact of occludin on the NFκB/SIRT pathway was evaluated. Both occludin depletion (Fig. 2A) and HIV infection (Fig. 2B) resulted in decreased SIRT-1 expression. The NADH-dependent nuclear translocation of CtBP1 has been associated to decreased SIRT-1 expression (18), the subcellular localization of CtBP1 was explored. Decreased cytoplasmic and increased nuclear localization of CtBP1 was found in the occludin-deficient pericytes (Fig. 2C, D) and in their HIV-infected counterparts (Fig. 2E, F).
Figure 2.
Occludin levels correlate with SIRT-1 expression and nuclear translocation of its repressor, CtBP1. A) SIRT-1 expression in noninfected pericytes treated with siRNA against occludin (OCC−) or SCR-treated or WT cells 48 h after silencing. P vs. WT; *P vs. SCR. B) SIRT-1 expression in WT pericytes 48 after HIV infection. C) Confocal microscopy images of CtBP1 immunolocalization (green) in pericytes treated with anti-occludin siRNA as in (A). Nuclear chromatin was counterstained with DRAQ5 (red). D) Magnified cell nuclei of pericyte cultures treated as in (C) showing nuclear immunolocalization of CtBP1 (cyan); chromatin was counterstained with DRAQ5. E) Confocal microscopy images of CtBP1 and nuclear chromatin in pericytes treated with siRNA, as in (C), 48 h after HIV infection. F) Magnified cell nuclei showing CtBP1 and chromatin staining in pericyte cultures treated as in (E), 48 h after infection. SIRT-1 expression in (A) and (B) was quantitated by SPRICE. Box-and-whisker graphs represent the range and the 75th, 50th, and 25th percentiles.
We next evaluated the phosphorylation state of SIRT-1 as a function of occludin expression and HIV infection by measuring phosphorylation at Ser47 which, together with phosphorylated Ser27 and Ser530, results in enhanced deacetylase activity (19). A decrease in phosphorylated Ser47 SIRT-1 was found in occludin-deficient (Fig. 3A) and HIV-infected (Fig. 3B) pericytes. To confirm that SIRT-1 levels are associated with occludin expression, total SIRT-1 and its Ser47 phosphorylation were quantified in occludin-overexpressing pericytes. The initial effect of HIV on occludin levels was replicated by treating pericytes with occludin siRNA for 12 h and then allowing the cells to recover by exchanging siRNA-containing medium for normal growth medium. This procedure resulted in occludin overexpression 48 h after medium was exchanged (Fig. 3C). In addition, the process was enhanced when SIRT-1 activity was repressed with HR73 (20 µM) for 12 h, indicating that SIRT-1 modulates occludin recovery in pericytes by providing a feedback mechanism. The levels of both total SIRT-1 and its Ser47 phosphorylated form were elevated in occludin-overexpressing pericytes (Fig. 3D, E).
Figure 3.
Occludin expression levels correlate with SIRT-1 activation. A) Expression of active SIRT-1 (phosphorylated at Ser47) in pericytes treated as in Fig. 2A 48 h after silencing. P vs. WT; *P vs. SCR. B) Active SIRT-1 expression in WT pericytes 48 h after HIV infection. C) Left: occludin levels in pericytes treated with siRNA as in (A); right: parallel cultures of pericytes similarly treated and later recovered (R) by exchanging siRNA-containing medium with normal growth medium 12 h after silencing. P vs. WT; *P vs. SCR. Confocal microscopy images show representative occludin immunostaining (green); nuclei were counterstained with DRAQ5 (red). Selected cultures were treated with the SIRT-1 inhibitor HR73 (20 µM for 12 h). D) SIRT-1 expression in occludin-overexpressing pericytes (labeled as in C). P vs. WT; *P vs. SCR. E) Active SIRT-1 expression in the same pericyte cultures as in (D). P vs. WT; *P vs. SCR. F) Occludin levels in HIV-infected occludin-recovered (R) pericytes (labeled as in C–E). G) SIRT-1 expression in occludin-recovered (OCC-R) pericytes 48 h after HIV infection. H) Active SIRT-1 expression in pericyte cultures used in (G). I) Transcriptional efficiency measured by GFP fluorescence in pericytes treated with siRNA as in (A) and recovered to induce occludin overexpression (OCC-R), as in (C) 48 h after infection. Expression of SIRT-1, active SIRT-1, and occludin was quantitated by SPRICE. P vs. WT; *P vs. SCR. Box-and-whisker plots represent the range and the 75th, 50th, and 25th percentiles. NI, noninfected.
To evaluate the effect of HIV infection on these events, we first induced pericytes to overexpress occludin and then infected them 24 h after siRNA recovery. Occludin levels were lower in pericytes infected for 48 h compared to those in the noninfected controls. However, in occludin-recovered/HIV-infected cells, occludin levels were still higher than in WT/noninfected pericytes (Fig. 3F). SIRT-1 expression and phosphorylation was then measured in the same recovered and infected pericytes. Under these conditions, SIRT-1 expression and the levels of its phosphorylated form were indistinguishable between infected and noninfected cells (Fig. 3G, 3H). To determine whether SIRT-1 recovery influences HIV transcription, we analyzed the same cultures spectrometrically to quantify GFP expression. Infection of occludin-recovered pericytes resulted in 3 times less HIV transcription than in WT cells (Fig. 3I).
Activation of SIRT-1 was also evaluated by measuring levels of acetylated NFκB-p65 (Lys310), which is deacetylated by SIRT-1. Both occludin deficiency and HIV infection elevated acetyl NFκB-p65. In addition, blocking SIRT-1 activity with HR73 (20 µM, 24 h) enhanced levels of acetyl NFκB-p65 that resembled those in occludin-deficient cells (Supplemental Fig. S1F, G).
Occludin is an NADH oxidase
To this point, our observations indicated that occludin modulates HIV infection by controlling the expression and activation of SIRT-1. On the other hand, SIRT-1 activation depends on NAD+, and its transcriptional repressor CtBP1 requires NADH to translocate into the nuclei. To clarify possible mechanisms underlying these effects in the onset of occludin expression, we studied the structure of the C-terminal domain of occludin. The crystal structure of the molecule has only been resolved for the CC domain (20), which accounts for the most distal third of the C-terminal domain (Fig. 4A). Therefore, we performed in silico modeling to predict the structure of the full C-terminal domain. This putative model contains a flexible hinge region (Fig. 4B, C, yellow) that partially encircles the CC domain (Fig. 4B, C, white), connecting it with the rest of the molecule (Fig. 4B, dark green). The ZO-1-binding site is located away from this hinge, which also contains the occludin phosphorylation switch. A putative NADH-binding site was identified in a pocket formed by complementation of the CC domain (Fig. 4B, C, blue) with the hinge (Fig. 4B, C, green). The amino acid sequence corresponding to the modeled region is shown in Fig. 4D. Comparison of the predicted model with the known structures of several proteins indexed in structural databases indicated similarities with oxidoreductases that use NADH/NAD+ as a cosubstrate (Tables 1 and 2).
Figure 4.
Hybrid model of the C-terminal domain of occludin. A) Reference structure of occludin showing its membranal localization and domain composition. Numbers correspond to the amino acid positions. N, amino terminal; C, carboxyl terminal. B) The human amino acid occludin sequence 266–522 was used to generate its predicted structural model. The known crystal structure of its CC domain (aa 416–522) is solid white. The structure of a smaller region from the C-terminal domain, encompassing aa 323–522, was further refined. The CC domain is solid white. The region surrounding the CC domain forms a hinge (yellow) that is flexible. For structural reference, the location of the multiphosphorylation region (red) and Cys409 (dark blue) involved in occludin self-oligomerization are shown. In the same hinge region, a putative binding site for NADH was identified, formed by complementation of a small area of the CC domain (blue) and the hinge (green). The binding site for ZO-1 (magenta) is located opposite the predicted localization of the hinge. C) Detailed structure of the NADH binding pocket (color code similar to C). In this model, the binding pocket form 5 hydrogen bonds with NADH, involving the aa D333, R335 (twice), K433, and K444. D) Amino acid sequence of the fragment 266–522. Color code as in (B) and (C). Amino acids predicted to bind NADH are underlined.
TABLE 1.
Enzyme classification database similarities
Rank | TM score | Identity (%) | Coverage (%) | PDB chain | EC number |
---|---|---|---|---|---|
1 | 0.4590 | 9 | 71 | 2cxi(A) | 6.1.1.20 |
2 | 0.4995 | 7 | 80 | 2fkn(B) | 4.2.1.49 |
3 | 0.4987 | 7 | 80 | 2fkn(A) | 4.2.1.49 |
4 | 0.3821 | 12 | 67 | 2gqd(A) | 2.3.1.179 |
5 | 0.4754 | 8 | 73 | 1s4f(A) | 2.7.7.48 |
Top proteins with structural similarities to the predicted occludin C-terminal domain model. EC, enzyme classification; PDB, Research Collaboratory for Structural Bioinformatics protein data bank structure; TM, template modeling score (higher score represents better similarity).
TABLE 2.
Gene ontology vocabulary database similarities
Rank | TM score | Identity (%) | Coverage (%) | PDB chain |
---|---|---|---|---|
1 | 0.7344 | 7 | 86 | 1zax(A) |
2 | 0.3524 | 13 | 59 | 1mlw(A) |
3 | 0.3686 | 12 | 61 | 2qk1(A) |
4 | 0.4096 | 11 | 63 | 2yxz(D) |
5 | 0.3699 | 14 | 52 | 1vmb(A) |
6 | 0.3765 | 10 | 71 | 2d0v(I) |
7 | 0.4375 | 9 | 70 | 2e3t(B) |
8 | 0.4666 | 8 | 77 | 1i19(B) |
9 | 0.4999 | 7 | 83 | 2fkn(D) |
10 | 0.4275 | 9 | 77 | 3c8y(A) |
Top proteins with structural similarities to the predicted occludin C-terminal model. PDB, Research Collaboratory for Structural Bioinformatics protein data bank structure; TM, template modeling score (higher score represents better similarity).
Similar modeling studies of intracellular domains of other TJ molecules revealed that the N-terminal domain of MARVEL D3 and the C-terminal domain of tricellulin, but not of claudin-5, share some structural similarities to other enzymes (lipases and DNA mismatch repair proteins, respectively; Supplemental Tables S1, S2), but not oxidoreductases. MARVEL D3 and tricellulin belong to the same protein family as occludin, implying a different evolutionary role for the members of this family of proteins. The intracellular domains of claudins are sufficiently small to prevent the formation of the structural foldings usually associated with enzymes.
To biochemically characterize occludin-mediated NADH and NAD+ interconversion, we incubated an NADH solution with the full C-terminal domain of occludin, originally expressed and purified from HEK-293 cells. Light-absorbance spectroscopy showed an increased NAD+:NADH ratio of the solution upon incubation with occludin, but not with occludin-free buffer or heat-denatured (100°C) occludin (Fig. 5A, J). In our experimental conditions (NADH 50–350 µM, 38°C, pH 7.3, with Ca++ and Mg++), occludin converted NADH into NAD+ at a maximum rate (Vmax) of 499.6 nmol/s with a Michaelis constant (Km) of 97 µM (Fig. 5B). Because our model suggested an NADH-binding pocket formed by complementation of the CC domain with the hinge, we explored whether the isolated CC domain of occludin also has effects on NADH/NAD+ conversion. Addition of the CC domain of murine occludin (99% similar in sequence to human) to a series of NADH solutions increased the conversion rate to a Vmax of 30.91 µmol/s with a Km of 229.5 µM (Fig. 5C, J). Thus, the full C-terminal domain binds NADH with higher affinity than the CC domain; however, the CC domain can convert NADH to NAD+ at high rates. Therefore, it is likely that the hinge, besides complementing the binding pocket and increasing the affinity for NADH, regulates the catalytic rate of the enzyme.
Figure 5.
Occludin has intrinsic NADH oxidase activity. A) Light absorption spectra of the purified C-terminal domain of human occludin incubated with 100 µM NADH solution. The conversion of NADH to NAD+, induced by the C-terminal (CT) domain and measured at 1 min (red), was higher than the spontaneous conversion in the solution if treated with protein-free buffer (blue). Heating the CT domain of occludin to 100°C resulted in decreased NADH oxidase capacity. B) Michaelis-Menten plot of the kinetics of the NADH-to-NAD+ conversion induced by the CT domain of human occludin in NADH solutions of increasing concentrations. C) Michaelis-Menten plot of the kinetics of the NADH-to-NAD+ conversion induced by the CC domain of murine occludin. D) Conversion rate of NADH-to-NAD+ of 100 µM NADH solution treated with a protein-free buffer, the CT domain of murine occludin, a cell lysate of HEK-293 cells, or a cell lysate of HEK-293 cells plus the CT domain of murine occludin. E) Confocal microscopy images of NADH in living WT or occludin-overexpressing HEK-293 cells (OCC+). F) Spectrometric quantitation of NADH in living WT and OCC+ HEK-293 cells. G) Confocal microscopy images showing nuclear localization of CtBP1 (green; Alexa 405) in WT and OCC+ HEK-293 cells. Nuclear chromatin was counterstained with DRAQ5 (red). Arrows: an increase in cytosolic CtBP1 in OCC+ cells. Occludin was detected with Alexa 594 (blue), precluding fluorescence bleed-through from occludin to the CtBP1 detection channel. H) Quantitation of occludin by SPRICE (left) and fluorescence intensity quantitation of CtBP1 colocalizing with heterochromatin (right) in OCC+ and WT HEK-293 cells. I) Cellular content of NADH measured in living primary human brain capillary pericytes treated with siRNA to silence occludin (OCC−), SCR, or WT. P vs. WT; *P vs. SCR. J) The regions of the CT domain of occludin used in (A–D), indicated in black. Numbers indicate amino acid positions. CT, full C-terminal domain encompassing the aa 266–522. Box-and-whisker graphs represent the range and the 75th, 50th, and 25th percentiles.
To explore the role of occludin as an oxidase in a cellular context, we added a lysate of HEK-293 cells to a NADH solution, resulting in an elevated NAD+:NADH ratio. This ratio increased further upon addition of the murine C-terminal domain, indicating enhanced NAD+ production (Fig. 5D). These findings were then validated in living cells. Biphotonic confocal microscopy evidenced a decrease in NADH content in living HEK-293 cells transiently overexpressing full-length occludin (Fig. 5E). Similarly, fluorescence spectroscopy showed a decrease in NADH content in living HEK-293 cells that overexpressed full-length occludin (Fig. 5F). These changes correlated functionally with nuclear translocation of CtBP1. Confocal microscopy of occludin-overexpressing HEK-293 cells showed increased cytosolic levels and less nuclear localization of CtBP1 than was observed in WT controls (Fig. 5G). Quantitation of occludin and CtBP1 evidenced a 3-fold increase in occludin expression that correlated with an ∼3-fold decrease in nuclear CtBP1 (Fig. 5H). To evaluate whether the occludin-induced changes in NADH levels found in HEK-293 cells also occur in pericytes, we measured NADH content in occludin-deficient pericytes. Occludin depletion resulted in enhanced cellular NADH levels when compared to WT cells (Fig. 5I). These observations indicated that the changes in subcellular localization of CtBP1 in pericytes (see Fig. 2C–F) were indeed caused by changes in cellular levels of NADH regulated by occludin levels. Variation in NADH levels correlated proportionally with variation in cellular flavin adenine dinucleotide FAD+ levels, indicating that the change in NADH/NAD+ ratio subsequent to alterations in occludin levels, did not alter the cellular redox ratio calculated as FAD+:NADH+FAD+ (Supplemental Fig. S3).
Correlating with the enzymatic role of occludin C-terminal domain, occludin immunoblot analysis in pericyte lysates indicated relatively low abundance of the canonical 65 kDa isoform 1 (typical epithelial occludin), but robust expression of isoforms 4 and 5 (31 and 23 kDa, respectively) (Supplemental Fig. S3H).
HIV replication correlates negatively with occludin levels in human macrophages
In the last series of experiments, we investigated to determine whether the observed regulatory effects of occludin on HIV transcription could be reproduced in HEK-293 cells and then in monocytes/macrophages, which share functional immunologic similarities with pericytes. Confocal microscopy and fluorescence spectroscopy of HEK-293 cells infected with HIV for 48 h revealed that occludin overexpression reduced HIV transcription to approximately half, compared with similarly infected WT controls (Fig. 6A, green signal; 6B). In addition, depletion of occludin resulted in a 3-fold increase in HIV transcription 48 h after infection of terminally differentiated U-937 monocytes (Fig. 6C), mimicking the results observed in HEK-292 cells and pericytes. We further explored these events in human primary macrophages differentiated from peripheral blood monocytes. Occludin depletion in noninfected macrophages resulted in augmented nuclear localization of CtBP1. HIV infection increased nuclear levels of CtBP1 48 h after infection in WT macrophages, an effect that was not further influenced by occludin depletion (Fig. 6D). Occludin depletion in human macrophages resulted in enhanced HIV transcription, as shown by confocal microscopy (Fig. 6E) and fluorescence spectroscopy (Fig. 6F), fully reproducing our observations in pericytes, HEK-293 cells, and U-937 monocytes.
Figure 6.
HIV replication correlates negatively with occludin levels in HEK-293 cells, monocytes, and human macrophages. A) Confocal microscopy images of HEK-293 cells overexpressing occludin (OCC+) or WT cells infected with HIV for 48 h. Green: viral GFP expression; blue: nuclear chromatin counterstained with DRAQ5; and red: occludin. B) Fluorescence spectrometry quantitation of HIV from media of the same HEK-293 cultures shown in (A). C) Similar quantitation of HIV from the supernatants of occludin-depleted U-937 monocytes treated with siRNA against occludin (OCC−), SCR, or WT, 48 h after infection. P vs. WT; *P vs. SCR. D) Confocal microscopy of human macrophages immunostained against occludin (red) and counterstained with DRAQ5 (blue nucleus) treated with siRNA against occludin (OCC−), SCR, or WT. Occludin content was quantified by SPRICE. P vs. WT; *P vs. SCR. Right: confocal microscopy images showing nuclear immunolocalization of CtBP1 48 h after HIV infection. NI, noninfected. E) Confocal microscopy images of HIV-infected human macrophages treated and infected with HIV as in (D). Green: viral intrinsic fluorescence; nuclear chromatin was counterstained with DRAQ5. F) Fluorescence spectrometry quantitation of HIV from supernatants of macrophages treated and infected with HIV, as in (D). P vs. WT; *P vs. SCR. Graphs show the range and the 75th, 50th, and 25th percentiles.
DISCUSSION
Although damage to the BBB occurs prominently during HIV infection and may contribute to the development of neurocognitive disorders, the direct interactions of HIV with cells of the neurovascular unit remain poorly understood. HIV has been found in brain capillary endothelial cells (21), but their direct and productive infection is still a matter for debate. HIV also infects astrocytes, even though they lack the CD4 receptor (22). Because of their heterogeneous nature and the lack of pericyte-specific cell markers, identification of HIV in BBB pericytes in brain tissue samples has not been properly confirmed in situ. Nevertheless, in vitro studies with primary human brain capillary pericytes indicated that they are CD4+ and can be directly infected by R5 (CCR5-tropic), X4 (CXCR4-tropic) and R5/X4 HIV variants (6). Although BBB pericytes are resistant to HIV’s cytopathic effects, surviving infection and permitting only low viral replication rates, they release IL-6, which results in functional alterations of endothelial cell TJ proteins (6). BBB pericytes are also capable of phagocytosis, express the macrophage markers ED-2 and CD11-b, and migrate in response to inflammatory mediators (2). They have the capacity to redifferentiate into glial lineages (23), suggesting that they are indeed targeted by HIV in vivo and potentially form viral reservoirs in the brain. This idea is partially supported by our results, showing that mild increases in viral inocula result in large changes in transcriptional rates, but only limited increases in viral release, implying that far more virus is produced than pericytes release to the medium. Similarly, previous work has shown that infected macrophages produce virions that are stored intracellularly, retaining infectivity for extended intervals even when viral transcription has stopped (24). Additional work, however, is needed to properly clarify whether pericytes indeed form viral reservoirs.
The current study demonstrates that HIV infection in BBB pericytes results in a dual-stage response pattern during the first 4 d after infection. The first stage (initial 48 h), is characterized by increased viral replication, a decrease in occludin expression, and increased acetylation of NFκB-p65. These events are associated with augmented nuclear localization of CtBP1 and reduced expression and activation of SIRT-1, whose activity is needed to control NFκB-p65 acetylation. The second stage of infection occurs in the following 48 h and is characterized by decreased viral replication, recovery of occludin levels, decreased nuclear translocation of CtBP1, recovery of SIRT-1, and subsequent decreased NFκB-p65 acetylation (Fig. 7). We also found a correlation between these stages with the activities of p38-MAPK and ROCK (Supplemental Fig. S1), which is in agreement with observations that p38-MAPK activity is involved in HIV replication (25). These effects are unlikely to be driven by autocrine activation, as IFN-α, -β, and -γ levels showed no difference between infected and noninfected pericytes (Supplemental Fig. S1E). These findings are consistent with our observations that production of most inflammatory cytokines is unaltered in pericytes after HIV infection (6).
Figure 7.
HIV infection of brain capillary pericytes triggers a biphasic metabolic response. The first stage involves depletion in occludin by a pathway that involves p38-MAPK activity. Occludin depletion leads to decreased NAD+ generation and increased NADH availability. Increased NADH results in enhanced nuclear translocation of CtBP1, which induces decreased SIRT-1 expression. In addition, diminished NAD+ availability decreases SIRT-1 phosphorylation and activation. In these conditions, levels of acetylated (i.e., transcriptionally active) NFκB-p65 are elevated, enhancing HIV replication. Pericytes escape from this pathway by increasing occludin expression in a manner that requires ROCK activity, shifting the infectious cycle to a second stage characterized by occludin recovery with subsequent reduction in NADH and nuclear CtBP1 translocation, normalized SIRT-1 expression, and increased SIRT-1 activation, resulting from elevated NAD+ availability. Increased SIRT-1 deacetylates active NFκB, hindering efficient HIV replication.
It has been shown that HIV hijacks the SIRT-1 pathway to benefit its own replication (8, 10). Our novel findings show that such control is acquired by depleting cellular occludin. The influence of occludin on SIRT-1 is remarkable in the settings of HIV infection, as elevated occludin levels have a prevailing effect over the virus in controlling SIRT-1 activation. The mechanism of this control is also notable. We characterized occludin as an NADH oxidase and showed that cellular levels of NADH inversely correlated with the cellular content of occludin. One of the metabolic outcomes of this process is that the NADH-dependent nuclear translocation of CtBP-1 is reduced (18), allowing SIRT-1 activation, given that the process depends on NAD+ availability (26). The binding of CtBP-1 to the promoter of several other genes is also NADH dependent (27), raising the possibility that other genes are regulated in a CtBP-1-mediated mechanism upon occludin overexpression. Whereas an increase in occludin levels results in elevated SIRT-1 expression, enhanced occludin expression appears to be inhibited by SIRT-1, as blocking SIRT-1 activity increased occludin recovery in our in vitro model (Fig. 3C). This finding suggests that although occludin controls SIRT-1 expression and activation, SIRT-1 provides negative feedback by modulating occludin levels. However, it should be pointed out that there may be additional modulatory mechanisms in infected brains. For example, it was reported that upregulation of microRNA-142 leads to repression of SIRT-1 in simian immunodeficiency virus (SIV) encephalitis (28).
In silico modeling indicated that the NADH oxidase activity of occludin resides in its C-terminal domain. Pericytes express primarily isoforms 4 and 5 of occludin (Supplemental Fig. S3H), which have conserved the C-terminal domains, identical in primary structure to the C-terminal domain of isoform 1 (the predominant endothelial and epithelial form of occludin). This expression pattern is consistent with a low level of membrane-bound occludin and the absence of TJs in pericytes. Isoform 4 lacks the first 3 transmembrane domains (thus is devoid of any extracellular loops), and isoform 5 does not have any transmembrane domain. In contrast, endothelial cells abundantly express membrane-bound isoforms 2 and 3 (29), but have low expression levels of isoforms 4 and 5. We have identified the C-terminal domain to be involved in occludin oligomerization in a thiol-sensitive manner (15), suggesting that occludin oligomerization influences its oxidase activity. These observations are in agreement with early literature data that low cytosolic thiol levels increase HIV replication (30). Our biochemical data combined with the in silico findings demonstrate that the enzymatic activity is regulated by changes in occludin conformation that affect the spatial position of the CC domain and the hinge. Under our experimental conditions, occludin showed a Km of 97 µM for NADH, falling within the ranges of other redox enzymes, including NAD(P)H oxidases (31). In our model, the differential-multiphosphorylation region of occludin was located in an exposed and readily accessible area, suggesting that conformational changes influence its enzymatic activity. Nevertheless, additional investigations are necessary to fully understand the structure–function relationship of occludin, in particular in nonepithelial and endothelial cells.
It has been demonstrated that the HIV protein Nef associates with the NADPH oxidase complex (32) and that Tat increases intracellular oxidants, thus activating c-Jun kinase, which induces SIRT-1 phosphorylation via NADPH (33). In our current work, HIV also had the capacity to alter the cellular NADH/NAD+ balance by changing the cellular levels of occludin. However, even under the high NAD+/NADH imbalances caused by occludin loss, the cellular redox potential appeared to remain unchanged, according to the FAD+:NADH+FAD+ ratio (Supplemental Fig. S3), supporting the idea that the NADH–NAD+ conversion caused by occludin is a signaling mechanism rather than a redox control.
Fully correlating with its enzymatic function, capacity to control SIRT-1, and modulation of HIV transcription, occludin depletion in a monocytic cell line and human macrophages resulted in a 2-fold increase in viral replication rates 48 h after infection. These results further indicate a potential role for occludin as a target for pharmacological intervention to approach viral reservoirs in HIV-infected patients. However, the influence of occludin on SIRT-1 may have additional effects, as SIRT-1 can epigenetically regulate gene expression affecting a variety of cellular events (26, 34). SIRT-1 also acts in concert with AMPK, to reduce energy expenditure; promote ATP synthesis; influence carbohydrate, protein, and lipid metabolism; and coordinate mitochondrial biogenesis, cell growth, and autophagy (35). Similar to SIRT-1, AMPK is activated by NAD+, for which it has stronger affinity in low AMP levels (36).
In epithelial and endothelial cells, occludin has been considered a regulator of TJ expression and function. Our current work provides further understanding of this regulatory role, as occludin can control TJ gene expression in a CtBP1- and SIRT-1-mediated manner. These observations strongly suggest that occludin plays a role in general cell metabolism. Indeed, data analysis of the gene atlas of the protein-encoding transcriptome (37) shows that occludin mRNA expression is ubiquitous in most human tissues, but exists most abundantly in those with higher metabolic rates, such as skeletal muscle, liver, and thyroid gland cells, as well as cardiomyocytes. In contrast, occludin mRNA abundance in epithelial and endothelial cells (i.e., cells responsible for generation of tissue barriers, where occludin was first discovered) is surprisingly low. Several metabolic pathways are amplified in such a manner that, to elicit a large response, only a small proportion of trigger molecules need be activated. This phenomenon may explain why a relative low (10%) change in occludin expression after HIV infection in pericytes has larger functional consequences.
Altogether, our observations indicate that occludin has previously unsuspected properties capable of modulating HIV-1 transcription. This result acquires particular relevance in a clinical setting where HIV infection goes hand in hand with the effort of viral eradication from cellular reservoirs. By regulating the NFκB/SIRT-1 pathway, occludin presents novel therapeutic and preventive targets that could be pharmacologically exploited. Furthermore, our data hint of the possibility that patients who have inflammatory conditions resulting in decreased occludin expression (e.g., chronic pain, stroke, or diabetes) are at a higher risk of developing higher HIV titers if they become infected. Our findings also show for the first time that occludin has a defined physiologic role outside its canonical TJ environment and in nonepithelial and endothelial cells.
Supplementary Material
Acknowledgments
The authors thank Mr. N. Ramsingh (University of Miami Miller School of Medicine) for his assistance with MARVEL D3 modeling. This work was supported by the U.S. National Institutes of Health (NIH) Heart, Lung, and Blood Institute Grant HL126559; Institute of Drug Abuse Grants DA039576 and DA027569; and Institute of Mental Health Grants MH098891, MH63022, and MH072567 and by the Miami Center for AIDS Research funded by NIH Grant P30AI073961. The authors declare no conflicts of interest.
Glossary
- BBB
blood–brain barrier
- CC
coiled–coil
- CCR
chemokine receptor, c-c motif
- CD
cluster of differentiation
- CtBP1
C-terminal-binding protein-1
- CXC
chemokine receptor, CXC motif
- DRAQ
DNA/RNA-binding anthraquinone
- GFP
green fluorescent protein
- HEK
human embryonic kidney cells
- M-CSF
macrophage colony-stimulating factor
- PCD
Protein Data Bank
- scpm
scintillation counts per minute
- SCR
scrambled
- siRNA
small interfering RNA
- SIRT-1
sirtuin 1
- SPRICE
spectrally resolved in-cell ELISA
- TJ
tight junctions
- WT
wild-type
- YFP
yellow fluorescent protein
- ZO
zonula occludens
Footnotes
This article includes supplemental data. Please visit http://www.fasebj.org to obtain this information.
REFERENCES
- 1.Bondjers C., He L., Takemoto M., Norlin J., Asker N., Hellström M., Lindahl P., Betsholtz C. (2006) Microarray analysis of blood microvessels from PDGF-B and PDGF-Rbeta mutant mice identifies novel markers for brain pericytes. FASEB J. 20, 1703–1705 [DOI] [PubMed] [Google Scholar]
- 2.Dore-Duffy P. (2008) Pericytes: pluripotent cells of the blood brain barrier. Curr. Pharm. Des. 14, 1581–1593 [DOI] [PubMed] [Google Scholar]
- 3.Armulik A., Abramsson A., Betsholtz C. (2005) Endothelial/pericyte interactions. Circ. Res. 97, 512–523 [DOI] [PubMed] [Google Scholar]
- 4.Shimizu F., Sano Y., Maeda T., Abe M. A., Nakayama H., Takahashi R., Ueda M., Ohtsuki S., Terasaki T., Obinata M., Kanda T. (2008) Peripheral nerve pericytes originating from the blood-nerve barrier expresses tight junctional molecules and transporters as barrier-forming cells. J. Cell. Physiol. 217, 388–399 [DOI] [PubMed] [Google Scholar]
- 5.Guillemin G. J., Brew B. J. (2004) Microglia, macrophages, perivascular macrophages, and pericytes: a review of function and identification. J. Leukoc. Biol. 75, 388–397 [DOI] [PubMed] [Google Scholar]
- 6.Nakagawa S., Castro V., Toborek M. (2012) Infection of human pericytes by HIV-1 disrupts the integrity of the blood-brain barrier. J. Cell. Mol. Med. 16, 2950–2957 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.West M. J., Lowe A. D., Karn J. (2001) Activation of human immunodeficiency virus transcription in T cells revisited: NF-kappaB p65 stimulates transcriptional elongation. J. Virol. 75, 8524–8537 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Pagans S., Pedal A., North B. J., Kaehlcke K., Marshall B. L., Dorr A., Hetzer-Egger C., Henklein P., Frye R., McBurney M. W., Hruby H., Jung M., Verdin E., Ott M. (2005) SIRT1 regulates HIV transcription via Tat deacetylation. PLoS Biol. 3, e41 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Scuto A., Kirschbaum M., Buettner R., Kujawski M., Cermak J. M., Atadja P., Jove R. (2013) SIRT1 activation enhances HDAC inhibition-mediated upregulation of GADD45G by repressing the binding of NF-κB/STAT3 complex to its promoter in malignant lymphoid cells. Cell Death Dis. 4, e635 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Zhang H-S., Zhou Y., Wu M-R., Zhou H-S., Xu F. (2009) Resveratrol inhibited Tat-induced HIV-1 LTR transactivation via NAD(+)-dependent SIRT1 activity. Life Sci. 85, 484–489 [DOI] [PubMed] [Google Scholar]
- 11.Huang W., Eum S. Y., András I. E., Hennig B., Toborek M. (2009) PPARalpha and PPARgamma attenuate HIV-induced dysregulation of tight junction proteins by modulations of matrix metalloproteinase and proteasome activities. FASEB J. 23, 1596–1606 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Blasig I. E., Bellmann C., Cording J., Del Vecchio G., Zwanziger D., Huber O., Haseloff R. F. (2011) Occludin protein family: oxidative stress and reducing conditions. Antioxid. Redox Signal. 15, 1195–1219 [DOI] [PubMed] [Google Scholar]
- 13.Bal M. S., Castro V., Piontek J., Rueckert C., Walter J. K., Shymanets A., Kurig B., Haase H., Nürnberg B., Blasig I. E. (2012) The hinge region of the scaffolding protein of cell contacts, zonula occludens protein 1, regulates interacting with various signaling proteins. J. Cell. Biochem. 113, 934–945 [DOI] [PubMed] [Google Scholar]
- 14.Blasig I. E., Winkler L., Lassowski B., Mueller S. L., Zuleger N., Krause E., Krause G., Gast K., Kolbe M., Piontek J. (2006) On the self-association potential of transmembrane tight junction proteins. Cell. Mol. Life Sci. 63, 505–514 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Walter J. K., Castro V., Voss M., Gast K., Rueckert C., Piontek J., Blasig I. E. (2009) Redox-sensitivity of the dimerization of occludin. Cell. Mol. Life Sci. 66, 3655–3662 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Chan J. K., Greene W. C. (2012) Dynamic roles for NF-κB in HTLV-I and HIV-1 retroviral pathogenesis. Immunol. Rev. 246, 286–310 [DOI] [PubMed] [Google Scholar]
- 17.Yang H., Zhang W., Pan H., Feldser H. G., Lainez E., Miller C., Leung S., Zhong Z., Zhao H., Sweitzer S., Considine T., Riera T., Suri V., White B., Ellis J. L., Vlasuk G. P., Loh C. (2012) SIRT1 activators suppress inflammatory responses through promotion of p65 deacetylation and inhibition of NF-κB activity. PLoS One 7, e46364 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Valenta T., Lukas J., Doubravska L., Fafilek B., Korinek V. (2006) HIC1 attenuates Wnt signaling by recruitment of TCF-4 and β-catenin to the nuclear bodies. EMBO J. 11, 2326–2337 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Nasrin N., Kaushik V. K., Fortier E., Wall D., Pearson K. J., de Cabo R., Bordone L. (2009) JNK1 phosphorylates SIRT1 and promotes its enzymatic activity. PLoS One 4, e8414 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Li Y., Fanning A. S., Anderson J. M., Lavie A. (2005) Structure of the conserved cytoplasmic C-terminal domain of occludin: identification of the ZO-1 binding surface. J. Mol. Biol. 352, 151–164 [DOI] [PubMed] [Google Scholar]
- 21.Bagasra O., Lavi E., Bobroski L., Khalili K., Pestaner J. P., Tawadros R., Pomerantz R. J. (1996) Cellular reservoirs of HIV-1 in the central nervous system of infected individuals: identification by the combination of in situ polymerase chain reaction and immunohistochemistry. AIDS 10, 573–585 [DOI] [PubMed] [Google Scholar]
- 22.Luo X., He J. J. (2015) Cell-cell contact viral transfer contributes to HIV infection and persistence in astrocytes. J. Neurovirol. 21, 66–80 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Dore-Duffy P., Katychev A., Wang X., Van Buren E. (2006) CNS microvascular pericytes exhibit multipotential stem cell activity. J. Cereb. Blood Flow Metab. 26, 613–624 [DOI] [PubMed] [Google Scholar]
- 24.Sharova N., Swingler C., Sharkey M., Stevenson M. (2005) Macrophages archive HIV-1 virions for dissemination in trans. EMBO J. 24, 2481–2489 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Furler R. L., Uittenbogaart C. H. (2010) Signaling through the P38 and ERK pathways: a common link between HIV replication and the immune response. Immunol. Res. 48, 99–109 [DOI] [PubMed] [Google Scholar]
- 26.Yeung F., Hoberg J. E., Ramsey C. S., Keller M. D., Jones D. R., Frye R. A., Mayo M. W. (2004) Modulation of NF-kappaB-dependent transcription and cell survival by the SIRT1 deacetylase. EMBO J. 23, 2369–2380 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Deng Y., Liu J., Han G., Lu S. L., Wang S. Y., Malkoski S., Tan A. C., Deng C., Wang X. J., Zhang Q. (2010) Redox-dependent Brca1 transcriptional regulation by an NADH-sensor CtBP1. Oncogene 29, 6603–6608 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Chaudhuri A. D., Yelamanchili S. V., Marcondes M. C., Fox H. S. (2013) Up-regulation of microRNA-142 in simian immunodeficiency virus encephalitis leads to repression of sirtuin1. FASEB J. 27, 3720–3729 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.McCaffrey G., Willis C. L., Staatz W. D., Nametz N., Quigley C. A., Hom S., Lochhead J. J., Davis T. P. (2009) Occludin oligomeric assemblies at tight junctions of the blood–brain barrier are altered by hypoxia and reoxygenation stress. J. Neurochem. 110, 58–71 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Staal F. J., Roederer M., Herzenberg L. A., Herzenberg L. A. (1990) Intracellular thiols regulate activation of nuclear factor kappa B and transcription of human immunodeficiency virus. Proc. Natl. Acad. Sci. USA 87, 9943–9947 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Nisimoto Y., Jackson H. M., Ogawa H., Kawahara T., Lambeth J. D. (2010) Constitutive NADPH-dependent electron transferase activity of the Nox4 dehydrogenase domain. Biochemistry 49, 2433–2442 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Salmen S., Colmenares M., Peterson D. L., Reyes E., Rosales J. D., Berrueta L. (2010) HIV-1 Nef associates with p22-phox, a component of the NADPH oxidase protein complex. Cell. Immunol. 263, 166–171 [DOI] [PubMed] [Google Scholar]
- 33.Gu Y., Wu R. F., Xu Y. C., Flores S. C., Terada L. S. (2001) HIV Tat activates c-Jun amino-terminal kinase through an oxidant-dependent mechanism. Virology 286, 62–71 [DOI] [PubMed] [Google Scholar]
- 34.Bordone L., Guarente L. (2005) Calorie restriction, SIRT1 and metabolism: understanding longevity. Nat. Rev. Mol. Cell Biol. 6, 298–305 [DOI] [PubMed] [Google Scholar]
- 35.Kahn B. B., Alquier T., Carling D., Hardie D. G. (2005) AMP-activated protein kinase: ancient energy gauge provides clues to modern understanding of metabolism. Cell Metab. 1, 15–25 [DOI] [PubMed] [Google Scholar]
- 36.Rafaeloff-Phail R., Ding L., Conner L., Yeh W. K., McClure D., Guo H., Emerson K., Brooks H. (2004) Biochemical regulation of mammalian AMP-activated protein kinase activity by NAD and NADH. J. Biol. Chem. 279, 52934–52939 [DOI] [PubMed] [Google Scholar]
- 37.Su A. I., Wiltshire T., Batalov S., Lapp H., Ching K. A., Block D., Zhang J., Soden R., Hayakawa M., Kreiman G., Cooke M. P., Walker J. R., Hogenesch J. B. (2004) A gene atlas of the mouse and human protein-encoding transcriptomes. Proc. Natl. Acad. Sci. USA 101, 6062–6067 [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.