Skip to main content
Erschienen in: Journal of Neuroinflammation 1/2018

Open Access 01.12.2018 | Research

Apolipoprotein E isoform dependently affects Tat-mediated HIV-1 LTR transactivation

verfasst von: Nabab Khan, Gaurav Datta, Jonathan D. Geiger, Xuesong Chen

Erschienen in: Journal of Neuroinflammation | Ausgabe 1/2018

Abstract

Background

Apolipoprotein E (ApoE) is the major carrier protein that mediates the transport and delivery of cholesterol and other lipids in the brain. Three isoforms of ApoE (ApoE2, ApoE3, ApoE4) exist in humans, and their relative expression levels impact HIV-1 infection, HIV-1/AIDS disease progression, and cognitive decline associated with HIV-1-associated neurocognitive disorder. Because HIV-1 Tat, a viral protein essential for HIV-1 replication, can bind to low-density lipoprotein receptor-related protein 1 (LRP1) that controls ApoE uptake in the brain, we determined the extent to which different isoforms of ApoE affected Tat-mediated HIV-1 LTR transactivation.

Methods

Using U87MG glioblastoma cells expressing LTR-driven luciferase, we determined the extent to which LRP1 as well as ApoE2, ApoE3, and ApoE4 affected Tat-mediated HIV-1 LTR transactivation.

Results

A specific LRP1 antagonist and siRNA knockdown of LRP1 both restricted significantly Tat-mediated LTR transactivation. Of the three ApoEs, ApoE4 was the least potent and effective at preventing HIV-1 Tat internalization and at decreasing Tat-mediated HIV-1 LTR transactivation. Further, Tat-mediated LTR transactivation was attenuated by an ApoE mimetic peptide, and ApoE4-induced restriction of Tat-mediated LTR transactivation was potentiated by an ApoE4 structure modulator that changes ApoE4 into an ApoE3-like phenotype.

Conclusions

These findings help explain observed differential effects of ApoEs on HIV-1 infectivity and the prevalence of HAND in people living with HIV-1 infection and suggest that ApoE mimetic peptides and ApoE4 structure modulator might be used as a therapeutic strategy against HIV-1 infection and associated neurocognitive disorders.

Background

Apolipoprotein E (ApoE), an apolipoprotein that mediates the transport and delivery of cholesterol and lipids through cell surface ApoE receptors [1], is highly expressed in the brain, and it plays a critical role in maintaining brain function [2, 3]. Three isoforms of ApoE with amino acid differences restricted to residues 112 and 158 have been identified in humans [1], and these differences affect the structure of ApoE isoforms, influence their ability to bind lipids and receptors [46], and contribute to disease pathogenesis. ApoE4, in addition to its involvement in Alzheimer’s disease [710] and cerebrovascular disease [11], is known to enhance HIV-1 infection [12], accelerate HIV-1 disease progression [12, 13], and increase the incidence of HIV-1-associated neurocognitive disorders (HAND) [14, 15].
Affecting 37 million people worldwide, HIV-1 virus can enter the brain within the first week of systemic infection [16, 17] after which it can harbor in CSF, perivascular macrophages, microglia, and astrocytes [18]. Although combined anti-retroviral therapy suppresses almost completely HIV-1 replication in plasma and CSF, reservoirs of HIV-1 exist and chronic low levels of neuroinflammation persist and appear to result in the 50% prevalence rate of HAND [19, 20]. Thus, additional strategies are needed to block viral reactivation to maintain HIV-1 in a state of prolonged silencing, to prevent disease progression, and to prevent HAND. One strategy might be the prevention of the transactivation of transcription protein (Tat) from activating HIV-1 replication because in the absence of Tat, transcription terminates prematurely due to inefficient elongation of the long terminal repeat (LTR) [2123].
Secreted from HIV-1-infected or HIV-transfected cells [24], Tat enters all CNS cells by endocytosis with the assistance of different receptors including low-density lipoprotein receptor-related protein 1 (LRP1), the major receptor for ApoE uptake in the brain; consequently, Tat is considered a key virulent factor of HIV-1 enhancing HIV infectivity and contributing to HIV/AIDS disease progression and the development of HAND [2527]. Further, of the three known human isoforms of ApoE, ApoE4 enhances HIV-1 infection and accelerates HIV-1 disease progression [12, 13, 28]. Accordingly, here, we tested the hypothesis that ApoE isoform dependently affects Tat-mediated HIV-1 LTR transactivation. Principally, we found that LRP1 was involved in Tat-mediated HIV-1 LTR transactivation, that ApoE4 was less effective than ApoE2 and ApoE3 in preventing HIV-1 Tat internalization and Tat-mediated HIV-1 LTR transactivation, that an ApoE mimetic peptide attenuated Tat-mediated LTR transactivation, and that an ApoE4 structure modulator enhanced the effectiveness of ApoE4 in blocking Tat-mediated HIV-1 LTR transactivation. These findings contribute to our understanding of ApoE and HIV-1 and suggest that ApoE mimetic peptides might be a therapeutic strategy against HIV-1 and HAND.

Methods

Cell culture

U87MG glioblastoma cells were cultured in 1× DMEM (Invitrogen) supplemented with 10% fetal calf serum and penicillin/streptomycin (Invitrogen). U87MG cells were stably transfected with luciferase reporter gene under the control of HIV-1 LTR promoter following neomycin (Sigma-Aldrich) selection pressure. These cells were provided generously by Dr. Lena Al-Harthi (Rush University, Chicago). ApoE2 and ApoE3 were purchased from Sigma-Aldrich, and ApoE4 from Abcam. Human plasma HDL were obtained from Kalen Biomedical and ApoE-deprived HDL3 (d = 1.125–1.21 g/ml) were prepared [29].

Luciferase reporter assay for Tat-mediated HIV-1 LTR transactivation

U87MG cells were seeded at a confluency level of about 50% (10,000 cells) on 96 well plates 1 day prior to being taken for experimentation. Cells were incubated with ApoE isoforms with or without HDL in the absence and presence of 2 μg/ml HIV-1 Tat (ABL Inc. and NIH AIDS program) protein and chloroquine (100 μM). Forty-eight hours post-incubation, luciferase activity assays (Promega) were performed and relative luminescence was quantified using a fluorometer/luminometer plate reader (Spectra MAX GEMINI EM). Similar protocols were followed using cells treated with HDL, LRP1 receptor-associated protein (RAP from American Research products), ApoE mimetic peptide (H-Gly-Arg-Leu-Val-Gln-Tyr-Arg-Gly-Glu-Val-Gln-Ala-Met-Leu-Gly-Gln-Ser-Thr-Glu-Glu-Leu-Arg-Val-Arg-Leu-Ala-Ser-His-Leu-Arg-Lys-Leu-Arg-Lys-Arg-Leu-Leu-Arg-Asp-OH from Anaspac Inc.), or ApoE4 structure corrector (PH002, Calbiochem).

RNA interference

U87MG cells were transfected with negative control siRNA (Ambion) and specific targeted LRP1 siRNA (sense: CAGACAAGAUUGAACGGAUtt and anti-sense: AUCCGUUCAAUCUUGUCUGTc from Ambion) by Jetprime reagent (Polyplus). Twenty-four hours post-transfection, cells were treated with HIV-1 Tat protein in the presence of chloroquine (100 μM) for 48 h and then taken for luciferase assay. Transfection efficiency was determined by protein levels of LRP1.

Immunoblotting

Cells were harvested and lysed in 1× RIPA lysis buffer (Thermofisher) plus 10 mM NaF, 1 mM Na3VO4, and Protease Inhibitor Cocktail (Sigma). After centrifugation (14,000×g for 10 min at 4 °C), supernatants were collected, and protein concentrations were determined with a DC protein assay (Bio-Rad). Proteins (10 μg) were separated by SDS-PAGE (12% gel) and transferred to polyvinylidene difluoride membranes (Millipore). The membranes were incubated overnight at 4 °C with antibodies against GAPDH (Abcam) and LRP1 (Abcam). The blots were developed with enhanced chemiluminescence, and bands were visualized and analyzed by our LI-COR Odyssey Fc Imaging System.

HIV-1 Tat internalization assay

Quantitative analysis of HIV-1 Tat internalization was performed using a method as described previously, but with minor modifications [30]. Cells plated on glass-bottom 35-mm2 tissue culture dishes were co-incubated with ApoE2, ApoE3, or ApoE4 (5,10 μg/ml) and FITC-labeled HIV-1 Tat (5 μg/ml; purchased from Rockland) for 90 min at 37 °C in the presence of chloroquine (100 μM). Cells were washed with an acid wash solution (0.2 M acetic acid, 0.5 M NaCl, pH 2.8) at 4 °C for 10 min and then washed with ice-cold PBS for 5 min to remove surface-bound HIV-1 Tat-FITC. Cells were fixed in 4% paraformaldehyde, and images were taken with a confocal laser-scanning microscope (Zeiss LSM800). All experiments were performed in triplicate. The average integrated intensity of HIV-1 Tat-FITC green signal per cell was calculated for each dish using ImageJ software.

Statistical analysis

All data were expressed as means ± SD. Statistical significance between two groups was analyzed with a Student’s t test, and statistical significance among multiple groups was analyzed with one-way ANOVA plus a Tukey post hoc test. p < 0.05 was considered to be statistically significant.

Results

LRP1 is involved in Tat-mediated HIV-1 LTR transactivation

For our studies, we measured Tat-mediated HIV-1 LTR transactivation using U87MG glioblastoma cells stably transfected with HIV-1 LTR-driven luciferase. With this system, concentration-dependent increases in Tat-mediated HIV-1 LTR transactivation were observed when exogenous HIV-1 Tat protein was co-applied at an optimal concentration of 100 μM chloroquine (Additional file 1: Figure S1), a weak base that neutralizes the acidic environment of lysosomes, prevents HIV-1 Tat degradation, and enhances nuclear delivery of HIV-1 Tat [26, 3133]. Thus, many factors related to HIV-1 infection could affect Tat-mediated LTR transactivation. Given that gp120 and TNFα have both been shown to neutralize endolysosome pH [3436], we determined the extent to which gp120 and TNFα affected Tat-mediated LTR transactivation in the absence of chloroquine, and neither affected Tat-mediated LTR transactivation (Additional file 1: Figure S2). However, we demonstrated that other endolysosome de-acidifying reagents acted in a similar way as chloroquine to enhance Tat-mediated LTR transactivation (Additional file 1: Figure S2), and these reagents include LYS01 (a free base) [37], bafilomycin (a specific vacuolar ATPase inhibitor), and KH7 (a selective soluble adenylyl cyclase inhibitor) [38]. We showed that 100 μM of chloroquine enhanced Tat-mediated LTR transactivation at Tat concentration as low as 0.5 μg/ml (35 nM) (Additional file 1: Figure S1). In the present study, we chose higher concentrations of Tat at 2 μg/ml (140 nM) to induce robust LTR transactivation so that we can demonstrate confidently whether ApoE or blocking LRP1 could affect Tat-mediated LTR transactivation. Using the above conditions (Tat at 2 μg/ml in the presence of 100 μM chloroquine), we next determined the mechanisms by which HIV-1 Tat enters the cells and the involvement of LRP1 in the induction of HIV-1 LTR transactivation in part because LRP1 is involved with HIV-1 Tat internalization [27]. First, using a specific LRP1 antagonist, receptor-associated protein (RAP), we demonstrated that RAP concentration dependently and significantly restricted Tat-mediated HIV-1 LTR transactivation (Fig. 1a). Next, we determined the extent to which siRNA knockdown of LRP1 affected Tat-mediated HIV-1 LTR transactivation. LRP1 protein expression levels were significantly decreased in cells transfected with LRP1 specific siRNA as compared to control (scrambled) siRNA (Fig. 1b). Further, the ability of HIV-1 Tat in conjunction with chloroquine to increase HIV-1 LTR transactivation was significantly attenuated in cells transfected with LRP1 specific siRNA as compared to control (scrambled) siRNA (Fig. 1c). Thus, siRNA knockdown of LRP1 blocked significantly [39] Tat-mediated LTR transactivation. Together, these data indicate that LRP1 is involved in Tat-mediated LTR transactivation in this system.

ApoE isoform dependently decreased Tat-mediated HIV-1 LTR transactivation in the presence of HDL

ApoE-rich lipoprotein is the major carrier for cholesterol transport in the brain and is a well-characterized ligand of LRP1. To determine the extent to which ApoE-rich lipoprotein affects Tat-mediated HIV-1 LTR transactivation, we needed to mimic the in situ structure and composition of ApoE-rich lipoproteins in the brain. ApoE-rich lipoproteins synthesized in situ in the brain is thought to be HDL-like particles composed of phospholipids and un-esterified cholesterol [3941]. We first determined the extent to which plasma-derived HDL (which contains low levels of ApoE) or ApoE-deprived HDL3 affects Tat-induced LTR transactivation, and we found neither plasma-derived HDL nor ApoE-deprived HDL3 at the concentrations of 50 μg/ml affected Tat-induced LTR transactivation (data not shown). To model the brain in situ ApoE-rich lipoproteins, we pre-incubated ApoE2, ApoE3, or ApoE4 with ApoE-deprived HDL3 with a fixed ratio of 0.4 for ApoE protein/HDL protein to form ApoE2-HDL, ApoE3-HDL, and ApoE4-HDL. This ratio was based on the findings that the phospholipid content of human plasma HDL is about 29% and the protein content of human plasma HDL is about 40%; thus, the ratio of phospholipids/protein content of HDL is about 0.7 [42]. Furthermore, the ApoE/phospholipid ratio in CSF is about 0.6 [43]. Therefore, if one assumes that the phospholipid (HDL) content is similar in plasma and CSF, the calculated ratio of ApoE protein/HDL protein content in CSF is 0.42. Using this 0.4 ratio, we demonstrated that ApoE2-HDL (Fig. 2a) and ApoE3-HDL (Fig. 2b), significantly and concentration dependently restricted Tat-mediated HIV-1 LTR transactivation. In contrast, ApoE4-HDL only restricted Tat-mediated HIV-1 LTR transactivation at the two highest concentrations of tested (Fig. 2c). ApoE4-HDL was less potent and less effective than were ApoE2-HDL and ApoE3-HDL at restricting HIV-1 Tat-mediated HIV-1 LTR transactivation (Fig. 2d); calculated IC50 values were 0.08 μg/ml for ApoE2-HDL, 0.03 μg/ml for ApoE3-HDL, and 10.0 μg/ml for ApoE4-HDL. Because ApoE2, ApoE3, and ApoE4 were all purified recombinant proteins and may contain endotoxin, the presence of endotoxin in these recombinant proteins may affect Tat-induced HIV-1 LTR transactivation. Thus, we further determined the extent to which endotoxin affects HIV-1 LTR transactivation. We found that endotoxin did not have any effect on Tat-mediated HIV-1 LTR transactivation in U87MG cells (Additional file 1: Figure S3).

ApoE isoform dependently decreased Tat-mediated HIV-1 LTR transactivation in the absence of HDL

We have shown that ApoE isoform dependently affected Tat-mediated HIV-1 LTR transactivation in the presence of HDL and that HDL alone does not affect Tat-mediated HIV-1 LTR transactivation. Although we did not determine the extent to which ApoE-HDL affects HIV-1 infection, our finding seems contradict with a previous report that non-lipidated ApoE4 at 10 μg/ml enhances HIV-1 infection in vitro [12]. Thus, we determined the extent to which ApoE isoforms in the absence of HDL-affected Tat-mediated HIV-1 LTR transactivation. We demonstrated that, in the absence of HDL, ApoE2 (Fig. 3a) and ApoE3 (Fig. 3b), but not ApoE4 (Fig. 3c) concentration dependently restricted Tat-mediated HIV-1 LTR transactivation. By comparing results of the effects of ApoEs on Tat-mediated HIV-1 LTR transactivation in the absence or presence of HDL, it was clear that ApoE2 (Fig. 3d), ApoE3 (Fig. 3e), and ApoE4 (Fig. 3f) were all more potent and effective at restricting Tat-mediated HIV-1 LTR transactivation when tested in the presence of HDL (a fixed ratio of 0.4 for ApoE protein/HDL protein); IC50 values for ApoE2 were 0.08 μg/ml in the presence of HDL and 2.2 μg/ml in the absence of HDL, and for ApoE3 were 0.03 μg/ml in the presence of HDL and 1.4 μg/ml in the absence of HDL. In the presence of HDL, ApoE4 at the two highest concentrations slightly restricted Tat-mediated HIV-1 LTR transactivation; however, ApoE4 concentration dependently enhanced Tat-mediated HIV-1 LTR transactivation in the absence of HDL (Fig. 3f). Thus, our finding is consistent with the previous report that non-lipidated ApoE4 enhances HIV-1 infection [12].

ApoE isoform dependently restricted HIV-1 tat internalization

ApoE and HIV-1 Tat are competitive ligands for LRP1, and ApoE can restrict HIV-1 Tat internalization (26). Accordingly, we next determined the extent to which ApoE2, ApoE3, and ApoE4 affected internalization of HIV-1 Tat. Control cells (Fig. 4a) and cells treated with ApoE4 at 5 and 10 μg/ml (Fig. 4a) exhibited a typical punctate staining pattern for internalized FITC-labeled HIV-1 Tat. ApoE2 at 5 and 10 μg/ml (Fig. 4a), and ApoE3 at 5 and 10 μg/ml (Fig. 4a) displayed less punctate staining of internalized FITC-labeled HIV-1 Tat as compared to control cells. When fluorescence intensity was quantified, FITC-labeled HIV-1 Tat internalization was decreased significantly and concentration dependently for ApoE2 and ApoE3 (Fig. 4b). In contrast, ApoE4 was ineffective in blocking Tat internalization (Fig. 4b).

Effects of ApoE mimetic peptide and ApoE4 structure corrector on Tat-mediated HIV-1 LTR transactivation

ApoE, a protein with 191 amino acids, has a receptor-binding region at residues 134–150 [44], and biological functions of ApoE are maintained with small fragments consisting of residues 141–150 [45]. We next determined the extent to which an ApoE mimetic peptide (residues 131–169) could affect Tat-mediated HIV-1 LTR transactivation. The ApoE mimetic peptide concentration dependently restricted Tat-mediated HIV-1 LTR transactivation (Fig. 5a). Small-molecule structure correctors (ApoE4 structure correctors) have been shown to modify the structure of ApoE4 to be more of a ApoE3-like phenotype [46]. Using the ApoE4 structure corrector at concentrations ranging from 2.5 to 10 μg/ml, we found that the ApoE4 structure corrector significantly and concentration dependently enhanced the efficiency of ApoE4 in restricting Tat-mediated LTR transactivation (Fig. 5b).

Discussion

Of the three known human isoforms of ApoE, ApoE4 enhances HIV-1 infection and accelerates HIV-1 disease progression [12, 13, 28]. HIV-1 Tat is an HIV-1 viral protein that is essential for HIV-1 replication. HIV-1 Tat is also a ligand for low-density lipoprotein receptor-related protein 1 (LRP1) and as such can compete for LRP1 binding with ApoEs and can control ApoE-cholesterol uptake. Accordingly, we determined the extent to which different isoforms of ApoE affected Tat-mediated HIV-1 LTR transactivation. Using U87MG glioblastoma cells expressing LTR-driven luciferase, the principal finding of our studies were (1) that a specific LRP1 antagonist, receptor-associated protein, and siRNA knockdown of LRP1 both restricted significantly Tat-mediated LTR transactivation, (2) that of the three ApoEs, ApoE4 was the least potent and effective at preventing HIV-1 Tat internalization and decreasing Tat-mediated HIV-1 LTR transactivation, (3) that Tat-mediated LTR transactivation was attenuated by an ApoE mimetic peptide, and (4) that ApoE4-induced restriction of Tat-mediated LTR transactivation was potentiated by an ApoE4 structure modulator that changes ApoE4 into an ApoE3-like phenotype.
Resting memory CD4+ T cells in the blood are the best characterized cellular HIV-1 reservoirs [47]. However, there are other sites of HIV persistence in the body. The brain is unique in terms of its “immune privileged” status, and the presence of the blood-brain barrier limits efficacious access to anti-retroviral drugs, and evidence does suggest that the brain is a viral reservoir for HIV-1 [48, 49]. Indeed, HIV-1 enters the brain within the first week of systemic infection and targets perivascular macrophages, microglia, and astrocytes [16, 17], and HIV DNA has been detected in the brains of HIV+ individuals whose infection was controlled by combined anti-retroviral therapy [50], and partial integrated HIV-1 provirus has been detected in the brain cells [5053]. In the brain, macrophages, microglia, and astrocytes can be infected by HIV-1. However, HIV-1 infection in macrophages or microglia is often productive while infection in astrocytes is less productive [54]. It is not clear what causes such a nuance. It is reported that endocytosis is involved in HIV-1 entry in both macrophages [54, 55] and astrocytes [54], and different set of receptors are used for HIV-1 entry in macrophages or astrocytes, which could partially account for the restrictive nature of HIV-1 infection in astrocytes. It has been reported that in HIV-1-infected brain, up to 19% astrocytes carry HIV-1 DNA [56, 57] and because of their abundance astrocytes could be an important viral reservoir in brain. The existence of such viral reservoirs that act as sanctuaries for HIV-1 have made the complete eradication of HIV-1 extremely challenging [50, 51, 57]. The HIV-1 protein Tat represents an attractive target for HIV-1 eradication of HIV-1 because HIV-1 Tat is essential for viral transcription [2123] and because brain levels of Tat remains elevated even though HIV-1 virus is below detectable levels [58]. HIV-1 Tat is secreted from HIV-1 infected cells and can bind to cell surface receptors including LRP1, heparin sulfate proteoglycan, CD26, and CXCR4. HIV-1 Tat enters all CNS cells via receptor-mediated endocytosis [25, 27, 31, 59] and following its internalization into endolysosomes, HIV-1 Tat enters the nucleus and activates the HIV-1 LTR promoter [26, 60, 61]. The importance of HIV-1 Tat to HIV-1 replication is further highlighted by findings that in the absence of HIV-1 Tat, HIV-1 LTR transcription terminates prematurely due to inefficient elongation [2123].
Currently, local concentrations of Tat in HIV-1-infected tissues are not known. In Tat-transfected cell lines, nanomolar concentrations of Tat were detected in HIV-1-infected cells [61], sera of HIV-1-infected individuals [62, 63], and in CSF of HIV-1-infected individuals [58]. However, even those levels are likely to underestimate Tat concentrations in HIV-1-infected tissues [64] because Tat is easily denatured and oxidized [65, 66], and it can be sequestered by endogenous anti-Tat antibodies [67, 68] and/or by glycosaminoglycans [63]. Tat protein is present in the brain tissue, as evidenced by the demonstration of Tat reactivity in post-mortem brain tissue of HIV+ individuals [58] and in HIV-infected macaques [27]. Such brain Tat could be secreted from infiltrating T cells and mononuclear cells [58] and infected perivascular microphage, microglia cell, and astrocytes [16, 17]. While in situ brain levels of Tat are unknown, nanomolar concentrations of Tat were detected in CSF of HIV-infected individuals [58]. Tat was detected in the CSF of only three of eight HIV-infected individuals [58], and this may be due to the non-uniformity of HIV-1 infection, and the fact that protein levels in CSF are much lower compared to protein levels in brain interstitial fluid. Even though Tat may not be measurable in some cases [69], it is clearly present because it is essential for viral replication [2123, 26, 60, 61] and Tat transcripts are produced earlier and in greater quantities in infected cells than are p24 or gp41 [70, 71]. This should not be viewed as unusual because Tat has yet to be measured in human brain or for that matter in the brains of Tat-transgenic mice. In addition, as shown for the well-accepted neurotoxins glutamate and kainate, alterations in the size of the extracellular space may change what might be a physiological concentration to a pathological concentration [72]. The size of the extracellular space varies from 10 to 20% in different brain regions and is dynamic; significant shrinkage of the space occurs with glial cell swelling and hypertrophy, as well as with changes in pH, potassium, and sodium. Thus, Tat levels in HIV-1-infected brain may vary dramatically depending on the factors listed above as well as Tat’s physical-chemical properties. Furthermore, Tat could accumulate in the brain over time even in ART-treated individuals. In support, protease inhibitors such as Darunavir do not prevent the formation of Tat in vitro [58], and it has been shown that antiviral therapy does not block the secretion of Tat in HIV-infected individuals [67]. Thus, it is not unreasonable for us to speculate that in situ brain levels of Tat could reach 2 μg/ml (140 nM) level as used in the present study, especially in local brain regions around HIV-1-infected cells.
Because Tat is easily denatured and oxidized [65, 66], higher (nM to μM) concentrations of Tat are often required when treating cells with Tat for prolonged periods. Typically, robust HIV-1 LTR transactivation requires high concentrations of exogenous HIV-1 Tat. In COS cells transiently transfected with HIV-1 LTR chloramphenicol acetyl transferase (CAT) reporter, Tat at concentrations greater than 0.5 μg/ml induced LTR transactivation [61], whereas in HL3T1 cells stably integrated with HIV-1 LTR CAT reporter and TZM-bl cells stably integrated with HIV-1 LTR-β-galactosidase reporter, Tat at concentrations of up to 1 μg/ml did not induce LTR transactivation [32]. In our system where U87MG was stably integrated with HIV-1 LTR luciferase reporter, Tat at concentrations of up to 8 μg/ml did not induce LTR transactivation. However, chloroquine, a well-known anti-malarial drug that de-acidifies endolysosomes significantly, enhances Tat-mediated HIV-1 LTR transactivation [26, 3133]. Consistent with these findings, we found that the optimal concentration of chloroquine to markedly enhance Tat-mediated HIV-1 LTR transactivation was 100 μM. We showed here that 100 μM of chloroquine enhanced Tat-mediated LTR transactivation at Tat concentration as low as 0.5 μg/ml (35 nM). In the present study, we chose higher concentrations of Tat at 2 μg/ml (140 nM) to induce robust LTR transactivation so that we can demonstrate confidently whether ApoE or blocking LRP1 could inhibit Tat-mediated LTR transactivation. Currently, the underlying mechanism whereby chloroquine enhances Tat-induced LTR transactivation is not known. It has been speculated that chloroquine-induced de-acidification of endolysosomes decreases Tat catabolism and enhances the nuclear delivery of Tat [32]. Consistent with this idea, lipofectamine-based transfection reagents, which recently have been shown to avoid lysosome degradation [73], has the ability to enhance exogenous Tat-induced LTR transactivation [31, 32]. Although chloroquine is not present in the brain, many other factors in HIV-1 infected brain could act similarly as chloroquine to induce endolysosome de-acidification and enhance Tat-mediated LTR transactivation. Given that gp120 and TNFα have both been shown to neutralize endolysosome pH [34, 36], we determined the extent to which gp120 and TNFα affected Tat-mediated LTR transactivation in the absence of chloroquine, but neither affected Tat-mediated LTR transactivation. However, we demonstrated that other endolysosome de-acidifying reagents act in a similar way as chloroquine to enhance Tat-mediated LTR transactivation, and these reagents include a known free base (LYS01) [37], a specific vacuolar ATPase inhibitor (bafilomycin), and a selective soluble adenylyl cyclase inhibitor (KH7) [38].
It is well known that Tat has the extraordinary ability to cross plasma membrane transporting a variety of macromolecules into a plethora of cell types. Tat can enter all CNS cells via endocytosis following interactions with specific cell surface proteins and receptors [25, 27, 31, 59]. Following endocytosis and internalization into endolysosomes, Tat can either harbor in endolysosomes or escape into the cytosol, where it transits to the nucleus and activates the HIV-1 LTR promoter [26, 60, 61]. It remains an open question on how Tat escapes endolysosomes. Interestingly, chloroquine has been shown to dramatically enhance Tat endolysosome escape [74]. However, the concept that chloroquine neutralizes endolysosome pH and prevents degradation of Tat in endolysosomes only partially explains why chloroquine enhances Tat endolysosome escape. Further mechanistic studies are warranted.
LRP1 is a large type I transmembrane protein, composed of a large 515-kDa N-terminal extracellular subunit and a non-covalently associated 85-kDa C- terminal transmembrane subunit [75]. The extracellular domain has four ligand-binding domains I–IV with 2, 8, 10, and 11 cysteine-rich complement-type repeats, respectively. The LRP1 ligand-binding domains II and IV are the major LRP1 binding regions interacting with a diverse array of approximately 40 structurally diverse ligands including ApoE [76]. The binding of ApoE to LRP1 leads to its endocytosis [77]. It is important to note that the core domain of Tat (amino acids 37–48) is directly involved in Tat interaction with LRP1 domains II, III, and IV [27]. Thus, ApoE could compete with Tat to bind LRP1. Using a stably transfected cell line with HIV-1 LTR-driven luciferase, we determined ApoE isoform-dependent effects on HIV-1 LTR transactivation. Tat-mediated HIV-1 LTR transactivation in U87MG cells was clearly mediated by LRP1 because the transactivation was decreased significantly by pharmacological inhibition using receptor-associated protein and by a siRNA knockdown strategy. In the absence of HDL, ApoE2 and ApoE3 concentration dependently blocked HIV-1 Tat internalization and attenuated Tat-mediated HIV-1 LTR transactivation; however, ApoE4 in the absence of HDL was inefficient in blocking HIV-1 Tat internalization and enhanced Tat-mediated HIV-1 LTR transactivation. Such a finding is consistent with previous report that non-lipidated ApoE4 at 10 μg/ml enhances HIV-1 infection in vitro [12]. Moreover, HDL binding to the ApoEs clearly increased the potency and effectiveness of all three ApoE isoforms in restricting Tat-mediated HIV-1 LTR transactivation. In the presence of HDL, the IC50 values for ApoE2 and ApoE3 to restrict Tat-induced HIV-1 LTR transactivation are 80 and 30 ng/ml, respectively. These IC50 values are close to reported ApoE concentrations (46 ng/ml for ApoE2 and 18 ng/ml for ApoE3) in mouse brain interstitial fluid [39], but much lower than reported human CSF ApoE concentrations (2.69–8 μg/ml) [43, 78]. Thus, we feel confident that our findings suggest that the brain in situ HDL-like ApoE lipoproteins exert restrictive effects on Tat-induced LTR transactivation.
The observed isoform-dependent effects of ApoE on Tat-mediated HIV-1 LTR transactivation could be due to the differences in their ability to bind lipids and receptors. Amino acid differences among the three isoforms of ApoE are restricted to residues 112 and 158 [1], and the change from cysteine to arginine affects their structure and their ability to bind lipids and receptors [4, 5]. Previous reports have shown that non-lipidated ApoE does bind to LRP1, but with much less affinity compared to lipdated ApoE [7982]. In addition, both structure and functional analyses have shown that ApoE3, but not ApoE4, preferentially binds and transports HDL, and the binding of HDL lipids opens the hinge region between N- and C-terminal domain of ApoEs allowing receptor-binding domain free access to ApoE receptors [6, 83, 84]. Indeed, ApoE4 has been shown to be less effective than ApoE2 or ApoE3 in binding LRP1 [82]. Our findings suggest that ApoE4 is less efficient than ApoE2 or ApoE3 in competing with Tat for LRP1 binding, blocking HIV-1 Tat internalization, and blocking Tat-mediated LTR transactivation.
The N-terminal of ApoE contains the putative receptor-binding site (residues 141–150) for LRP1 [44], and ApoE mimetic peptides containing the receptor-binding region exhibit biological functions of ApoE [45]. Here, we demonstrated that ApoE mimetic peptide attenuated Tat-mediated HIV-1 LTR transactivation. Thus, ApoE mimetic peptide might be used as a therapeutic strategy against HIV-1. Furthermore, because small-molecule structure correctors (ApoE4 structure correctors) modify ApoE4 structure to be more ApoE3-like [46], we tested the effects of an ApoE4 structure corrector and demonstrated that the ApoE4 structure corrector enhanced the efficiency of ApoE4 in restricting Tat-mediated HIV-1 LTR transactivation.

Conclusions

Our findings indicate that ApoE4 is less effective than ApoE2 or ApoE3 in restricting Tat-induced HIV-1 LTR transactivation in glioblastoma, ApoE mimetic peptide could be used as a therapeutic strategy against HIV-1 infection and associated neurocognitive disorders, and ApoE4 structure correctors could be used as a therapeutic strategy in HIV-1 positive people having the APOE4 allele. Future studies investigating ApoE4 structure correctors and their ability to affect HIV-1 infection in astrocytes and macrophage/microglia cells and disease progression are warranted.

Acknowledgements

We thank Dr. Lena Al-Harthi (Rush University, Chicago), who provided us with U87MG cells which were stably transfected with luciferase reporter gene under the control of HIV-1 LTR promoter.

Funding

This work was supported by P30GM103329, R01MH100972, and R01MH105329.

Availability of data and materials

Not applicable.
Not applicable.
Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Mahley RW. Apolipoprotein E: cholesterol transport protein with expanding role in cell biology. Science. 1988;240:622–30.CrossRefPubMed Mahley RW. Apolipoprotein E: cholesterol transport protein with expanding role in cell biology. Science. 1988;240:622–30.CrossRefPubMed
2.
Zurück zum Zitat Rebeck GW, Kindy M, LaDu MJ. Apolipoprotein E and Alzheimer’s disease: the protective effects of ApoE2 and E3. J Alzheimers Dis. 2002;4:145–54.CrossRefPubMed Rebeck GW, Kindy M, LaDu MJ. Apolipoprotein E and Alzheimer’s disease: the protective effects of ApoE2 and E3. J Alzheimers Dis. 2002;4:145–54.CrossRefPubMed
3.
Zurück zum Zitat Poirier J. Apolipoprotein E represents a potent gene-based therapeutic target for the treatment of sporadic Alzheimer’s disease. Alzheimers Dement. 2008;4:S91–7.CrossRefPubMed Poirier J. Apolipoprotein E represents a potent gene-based therapeutic target for the treatment of sporadic Alzheimer’s disease. Alzheimers Dement. 2008;4:S91–7.CrossRefPubMed
4.
Zurück zum Zitat Garai K, Baban B, Frieden C. Self-association and stability of the ApoE isoforms at low pH: implications for ApoE-lipid interactions. Biochemistry. 2011;50:6356–64.CrossRefPubMed Garai K, Baban B, Frieden C. Self-association and stability of the ApoE isoforms at low pH: implications for ApoE-lipid interactions. Biochemistry. 2011;50:6356–64.CrossRefPubMed
5.
Zurück zum Zitat Yamamoto T, Choi HW, Ryan RO. Apolipoeprotein E isoform-specific binding to the low-density lipoprotein receptor. Anal Biochem. 2008;372:222–6.CrossRefPubMed Yamamoto T, Choi HW, Ryan RO. Apolipoeprotein E isoform-specific binding to the low-density lipoprotein receptor. Anal Biochem. 2008;372:222–6.CrossRefPubMed
6.
Zurück zum Zitat Frieden C, Wang H, Ho CMW. A mechanism for lipid binding to apoE and the role of intrinsically disordered regions coupled to domain-domain interactions. Proc Natl Acad Sci U S A. 2017;114:6292–7.CrossRefPubMedPubMedCentral Frieden C, Wang H, Ho CMW. A mechanism for lipid binding to apoE and the role of intrinsically disordered regions coupled to domain-domain interactions. Proc Natl Acad Sci U S A. 2017;114:6292–7.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Reitz C, Rogaeva E, Foroud T, Farrer LA. Genetics and genomics of late-onset Alzheimer’s disease and its endophenotypes. Int J Alzheimers Dis. 2011;2011:284728.PubMedPubMedCentral Reitz C, Rogaeva E, Foroud T, Farrer LA. Genetics and genomics of late-onset Alzheimer’s disease and its endophenotypes. Int J Alzheimers Dis. 2011;2011:284728.PubMedPubMedCentral
8.
Zurück zum Zitat Farrer LA, Cupples LA, Haines JL, Hyman B, Kukull WA, Mayeux R, Myers RH, Pericak-Vance MA, Risch N, van Duijn CM. Effects of age, sex, and ethnicity on the association between apolipoprotein E genotype and Alzheimer disease. A meta-analysis. APOE and Alzheimer Disease Meta Analysis Consortium. JAMA. 1997;278:1349–56.CrossRefPubMed Farrer LA, Cupples LA, Haines JL, Hyman B, Kukull WA, Mayeux R, Myers RH, Pericak-Vance MA, Risch N, van Duijn CM. Effects of age, sex, and ethnicity on the association between apolipoprotein E genotype and Alzheimer disease. A meta-analysis. APOE and Alzheimer Disease Meta Analysis Consortium. JAMA. 1997;278:1349–56.CrossRefPubMed
9.
Zurück zum Zitat Harold D, Abraham R, Hollingworth P, Sims R, Gerrish A, Hamshere ML, Pahwa JS, Moskvina V, Dowzell K, Williams A, et al. Genome-wide association study identifies variants at CLU and PICALM associated with Alzheimer’s disease. Nat Genet. 2009;41:1088–93.CrossRefPubMedPubMedCentral Harold D, Abraham R, Hollingworth P, Sims R, Gerrish A, Hamshere ML, Pahwa JS, Moskvina V, Dowzell K, Williams A, et al. Genome-wide association study identifies variants at CLU and PICALM associated with Alzheimer’s disease. Nat Genet. 2009;41:1088–93.CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Lambert JC, Heath S, Even G, Campion D, Sleegers K, Hiltunen M, Combarros O, Zelenika D, Bullido MJ, Tavernier B, et al. Genome-wide association study identifies variants at CLU and CR1 associated with Alzheimer’s disease. Nat Genet. 2009;41:1094–9.CrossRefPubMed Lambert JC, Heath S, Even G, Campion D, Sleegers K, Hiltunen M, Combarros O, Zelenika D, Bullido MJ, Tavernier B, et al. Genome-wide association study identifies variants at CLU and CR1 associated with Alzheimer’s disease. Nat Genet. 2009;41:1094–9.CrossRefPubMed
11.
Zurück zum Zitat Frank A, Diez-Tejedor E, Bullido MJ, Valdivieso F, Barreiro P. APOE genotype in cerebrovascular disease and vascular dementia. J Neurol Sci. 2002;203-204:173–6.CrossRefPubMed Frank A, Diez-Tejedor E, Bullido MJ, Valdivieso F, Barreiro P. APOE genotype in cerebrovascular disease and vascular dementia. J Neurol Sci. 2002;203-204:173–6.CrossRefPubMed
12.
Zurück zum Zitat Burt TD, Agan BK, Marconi VC, He W, Kulkarni H, Mold JE, Cavrois M, Huang Y, Mahley RW, Dolan MJ, et al. Apolipoprotein (apo) E4 enhances HIV-1 cell entry in vitro, and the APOE epsilon4/epsilon4 genotype accelerates HIV disease progression. Proc Natl Acad Sci U S A. 2008;105:8718–23.CrossRefPubMedPubMedCentral Burt TD, Agan BK, Marconi VC, He W, Kulkarni H, Mold JE, Cavrois M, Huang Y, Mahley RW, Dolan MJ, et al. Apolipoprotein (apo) E4 enhances HIV-1 cell entry in vitro, and the APOE epsilon4/epsilon4 genotype accelerates HIV disease progression. Proc Natl Acad Sci U S A. 2008;105:8718–23.CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Corder E, Saunders A, Strittmatter W, Schmechel D, Gaskell P, Small G, Roses A, Haines J, Pericak-Vance M. Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer’s disease in late onset families. Science. 1993;261:921–3.CrossRefPubMed Corder E, Saunders A, Strittmatter W, Schmechel D, Gaskell P, Small G, Roses A, Haines J, Pericak-Vance M. Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer’s disease in late onset families. Science. 1993;261:921–3.CrossRefPubMed
14.
Zurück zum Zitat Saylor D, Dickens AM, Sacktor N, Haughey N, Slusher B, Pletnikov M, Mankowski JL, Brown A, Volsky DJ, McArthur JC. HIV-associated neurocognitive disorder––pathogenesis and prospects for treatment. Nat Rev Neurol. 2016;12:234–48.CrossRefPubMedPubMedCentral Saylor D, Dickens AM, Sacktor N, Haughey N, Slusher B, Pletnikov M, Mankowski JL, Brown A, Volsky DJ, McArthur JC. HIV-associated neurocognitive disorder––pathogenesis and prospects for treatment. Nat Rev Neurol. 2016;12:234–48.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Wendelken LA, Jahanshad N, Rosen HJ, Busovaca E, Allen I, Coppola G, Adams C, Rankin KP, Milanini B, Clifford K, et al. ApoE epsilon4 is associated with cognition, brain integrity, and atrophy in HIV over age 60. J Acquir Immune Defic Syndr. 2016;73:426–32.CrossRefPubMedPubMedCentral Wendelken LA, Jahanshad N, Rosen HJ, Busovaca E, Allen I, Coppola G, Adams C, Rankin KP, Milanini B, Clifford K, et al. ApoE epsilon4 is associated with cognition, brain integrity, and atrophy in HIV over age 60. J Acquir Immune Defic Syndr. 2016;73:426–32.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Spudich S, Gonzalez-Scarano F. HIV-1-related central nervous system disease: current issues in pathogenesis, diagnosis, and treatment. Cold Spring Harb Perspect Med. 2012;2:a007120.CrossRefPubMedPubMedCentral Spudich S, Gonzalez-Scarano F. HIV-1-related central nervous system disease: current issues in pathogenesis, diagnosis, and treatment. Cold Spring Harb Perspect Med. 2012;2:a007120.CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Valcour V, Chalermchai T, Sailasuta N, Marovich M, Lerdlum S, Suttichom D, Suwanwela NC, Jagodzinski L, Michael N, Spudich S, et al. Central nervous system viral invasion and inflammation during acute HIV infection. J Infect Dis. 2012;206:275–82.CrossRefPubMedPubMedCentral Valcour V, Chalermchai T, Sailasuta N, Marovich M, Lerdlum S, Suttichom D, Suwanwela NC, Jagodzinski L, Michael N, Spudich S, et al. Central nervous system viral invasion and inflammation during acute HIV infection. J Infect Dis. 2012;206:275–82.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Wong JK, Yukl SA. Tissue reservoirs of HIV. Curr Opin HIV AIDS. 2016;11:362–70. Wong JK, Yukl SA. Tissue reservoirs of HIV. Curr Opin HIV AIDS. 2016;11:362–70.
19.
Zurück zum Zitat Ellis RJ, Rosario D, Clifford DB, McArthur JC, Simpson D, Alexander T, Gelman BB, Vaida F, Collier A, Marra CM, et al. Continued high prevalence and adverse clinical impact of human immunodeficiency virus-associated sensory neuropathy in the era of combination antiretroviral therapy: the CHARTER study. Arch Neurol. 2010;67:552–8.CrossRefPubMedPubMedCentral Ellis RJ, Rosario D, Clifford DB, McArthur JC, Simpson D, Alexander T, Gelman BB, Vaida F, Collier A, Marra CM, et al. Continued high prevalence and adverse clinical impact of human immunodeficiency virus-associated sensory neuropathy in the era of combination antiretroviral therapy: the CHARTER study. Arch Neurol. 2010;67:552–8.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Heaton RK, Clifford DB, Franklin DR Jr, Woods SP, Ake C, Vaida F, Ellis RJ, Letendre SL, Marcotte TD, Atkinson JH, et al. HIV-associated neurocognitive disorders persist in the era of potent antiretroviral therapy: CHARTER study. Neurology. 2010;75:2087–96.CrossRefPubMedPubMedCentral Heaton RK, Clifford DB, Franklin DR Jr, Woods SP, Ake C, Vaida F, Ellis RJ, Letendre SL, Marcotte TD, Atkinson JH, et al. HIV-associated neurocognitive disorders persist in the era of potent antiretroviral therapy: CHARTER study. Neurology. 2010;75:2087–96.CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Romani B, Engelbrecht S, Glashoff RH. Functions of Tat: the versatile protein of human immunodeficiency virus type 1. J Gen Virol. 2010;91:1–12.CrossRefPubMed Romani B, Engelbrecht S, Glashoff RH. Functions of Tat: the versatile protein of human immunodeficiency virus type 1. J Gen Virol. 2010;91:1–12.CrossRefPubMed
22.
Zurück zum Zitat Jeang KT, Xiao H, Rich EA. Multifaceted activities of the HIV-1 transactivator of transcription, Tat. J Biol Chem. 1999;274:28837–40. Jeang KT, Xiao H, Rich EA. Multifaceted activities of the HIV-1 transactivator of transcription, Tat. J Biol Chem. 1999;274:28837–40.
23.
Zurück zum Zitat Clark E, Brenda N, Massimo C. Tat is a multifunctional viral protein that modulates cellular gene expression and functions. Oncotarget. 2017;8:27569–81.CrossRefPubMedPubMedCentral Clark E, Brenda N, Massimo C. Tat is a multifunctional viral protein that modulates cellular gene expression and functions. Oncotarget. 2017;8:27569–81.CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Agostini S, Ali H, Vardabasso C, Fittipaldi A, Tasciotti E, Cereseto A, Bugatti A, Rusnati M, Lusic M, Giacca M. Inhibition of non canonical HIV-1 Tat secretion through the cellular Na+, K+-ATPase blocks HIV-1 infection. EBioMedicine. 2017;21:170–181. Agostini S, Ali H, Vardabasso C, Fittipaldi A, Tasciotti E, Cereseto A, Bugatti A, Rusnati M, Lusic M, Giacca M. Inhibition of non canonical HIV-1 Tat secretion through the cellular Na+, K+-ATPase blocks HIV-1 infection. EBioMedicine. 2017;21:170–181.
25.
Zurück zum Zitat Debaisieux S, Rayne F, Yezid H, Beaumelle B. The ins and outs of HIV-1 Tat. Traffic. 2012;13:355–63.CrossRefPubMed Debaisieux S, Rayne F, Yezid H, Beaumelle B. The ins and outs of HIV-1 Tat. Traffic. 2012;13:355–63.CrossRefPubMed
26.
Zurück zum Zitat Vendeville A, Rayne F, Bonhoure A, Bettache N, Montcourrier P, Beaumelle B. HIV-1 Tat enters T cells using coated pits before translocating from acidified endosomes and eliciting biological responses. Mol Biol Cell. 2004;15:2347–60.CrossRefPubMedPubMedCentral Vendeville A, Rayne F, Bonhoure A, Bettache N, Montcourrier P, Beaumelle B. HIV-1 Tat enters T cells using coated pits before translocating from acidified endosomes and eliciting biological responses. Mol Biol Cell. 2004;15:2347–60.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Liu Y, Jones M, Hingtgen CM, Bu G, Laribee N, Tanzi RE, Moir RD, Nath A, He JJ. Uptake of HIV-1 tat protein mediated by low-density lipoprotein receptor-related protein disrupts the neuronal metabolic balance of the receptor ligands. Nat Med. 2000;6:1380–7.CrossRefPubMed Liu Y, Jones M, Hingtgen CM, Bu G, Laribee N, Tanzi RE, Moir RD, Nath A, He JJ. Uptake of HIV-1 tat protein mediated by low-density lipoprotein receptor-related protein disrupts the neuronal metabolic balance of the receptor ligands. Nat Med. 2000;6:1380–7.CrossRefPubMed
28.
Zurück zum Zitat Dobson CB, Sales SD, Hoggard P, Wozniak MA, Crutcher KA. The receptor-binding region of human apolipoprotein E has direct anti-infective activity. J Infect Dis. 2006;193:442–50.CrossRefPubMed Dobson CB, Sales SD, Hoggard P, Wozniak MA, Crutcher KA. The receptor-binding region of human apolipoprotein E has direct anti-infective activity. J Infect Dis. 2006;193:442–50.CrossRefPubMed
29.
Zurück zum Zitat Heeren J, Grewal T, Laatsch A, Rottke D, Rinninger F, Enrich C, Beisiegel U. Recycling of apoprotein E is associated with cholesterol efflux and high density lipoprotein internalization. J Biol Chem. 2003;278:14370–8.CrossRefPubMed Heeren J, Grewal T, Laatsch A, Rottke D, Rinninger F, Enrich C, Beisiegel U. Recycling of apoprotein E is associated with cholesterol efflux and high density lipoprotein internalization. J Biol Chem. 2003;278:14370–8.CrossRefPubMed
30.
Zurück zum Zitat Li S, Geiger NH, Soliman ML, Hui L, Geiger JD, Chen X. Caffeine, through adenosine A3 receptor-mediated actions, suppresses amyloid-beta protein precursor internalization and amyloid-beta generation. J Alzheimers Dis. 2015;47:73–83.CrossRefPubMedPubMedCentral Li S, Geiger NH, Soliman ML, Hui L, Geiger JD, Chen X. Caffeine, through adenosine A3 receptor-mediated actions, suppresses amyloid-beta protein precursor internalization and amyloid-beta generation. J Alzheimers Dis. 2015;47:73–83.CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Frankel AD, Pabo CO. Cellular uptake of the tat protein from human immunodeficiency virus. Cell. 1988;55:1189–93.CrossRefPubMed Frankel AD, Pabo CO. Cellular uptake of the tat protein from human immunodeficiency virus. Cell. 1988;55:1189–93.CrossRefPubMed
32.
Zurück zum Zitat Li GH, Li W, Mumper RJ, Nath A. Molecular mechanisms in the dramatic enhancement of HIV-1 Tat transduction by cationic liposomes. FASEB J. 2012;26:2824–34.CrossRefPubMedPubMedCentral Li GH, Li W, Mumper RJ, Nath A. Molecular mechanisms in the dramatic enhancement of HIV-1 Tat transduction by cationic liposomes. FASEB J. 2012;26:2824–34.CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Kolson DL, Collman R, Hrin R, Balliet JW, Laughlin M, McGann KA, Debouck C, Gonzalez-Scarano F. Human immunodeficiency virus type 1 Tat activity in human neuronal cells: uptake and trans-activation. J Gen Virol. 1994;75(Pt 8):1927–34.CrossRefPubMed Kolson DL, Collman R, Hrin R, Balliet JW, Laughlin M, McGann KA, Debouck C, Gonzalez-Scarano F. Human immunodeficiency virus type 1 Tat activity in human neuronal cells: uptake and trans-activation. J Gen Virol. 1994;75(Pt 8):1927–34.CrossRefPubMed
34.
Zurück zum Zitat Bae M, Patel N, Xu H, Lee M, Tominaga-Yamanaka K, Nath A, Geiger J, Gorospe M, Mattson MP, Haughey NJ. Activation of TRPML1 clears intraneuronal Abeta in preclinical models of HIV infection. J Neurosci. 2014;34:11485–503.CrossRefPubMedPubMedCentral Bae M, Patel N, Xu H, Lee M, Tominaga-Yamanaka K, Nath A, Geiger J, Gorospe M, Mattson MP, Haughey NJ. Activation of TRPML1 clears intraneuronal Abeta in preclinical models of HIV infection. J Neurosci. 2014;34:11485–503.CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat Hui L, Chen X, Haughey NJ, Geiger JD. Role of endolysosomes in HIV-1 Tat-induced neurotoxicity. ASN Neuro. 2012;4:243–52.CrossRefPubMed Hui L, Chen X, Haughey NJ, Geiger JD. Role of endolysosomes in HIV-1 Tat-induced neurotoxicity. ASN Neuro. 2012;4:243–52.CrossRefPubMed
36.
Zurück zum Zitat Nilsson C, Johansson U, Johansson AC, Kagedal K, Ollinger K. Cytosolic acidification and lysosomal alkalinization during TNF-alpha induced apoptosis in U937 cells. Apoptosis. 2006;11:1149–59.CrossRefPubMed Nilsson C, Johansson U, Johansson AC, Kagedal K, Ollinger K. Cytosolic acidification and lysosomal alkalinization during TNF-alpha induced apoptosis in U937 cells. Apoptosis. 2006;11:1149–59.CrossRefPubMed
37.
Zurück zum Zitat McAfee Q, Zhang Z, Samanta A, Levi SM, Ma XH, Piao S, Lynch JP, Uehara T, Sepulveda AR, Davis LE, et al. Autophagy inhibitor Lys05 has single-agent antitumor activity and reproduces the phenotype of a genetic autophagy deficiency. Proc Natl Acad Sci U S A. 2012;109:8253–8.CrossRefPubMedPubMedCentral McAfee Q, Zhang Z, Samanta A, Levi SM, Ma XH, Piao S, Lynch JP, Uehara T, Sepulveda AR, Davis LE, et al. Autophagy inhibitor Lys05 has single-agent antitumor activity and reproduces the phenotype of a genetic autophagy deficiency. Proc Natl Acad Sci U S A. 2012;109:8253–8.CrossRefPubMedPubMedCentral
38.
Zurück zum Zitat Rahman N, Ramos-Espiritu L, Milner TA, Buck J, Levin LR. Soluble adenylyl cyclase is essential for proper lysosomal acidification. J Gen Physiol. 2016;148:325–39.CrossRefPubMedPubMedCentral Rahman N, Ramos-Espiritu L, Milner TA, Buck J, Levin LR. Soluble adenylyl cyclase is essential for proper lysosomal acidification. J Gen Physiol. 2016;148:325–39.CrossRefPubMedPubMedCentral
39.
Zurück zum Zitat Ulrich JD, Burchett JM, Restivo JL, Schuler DR, Verghese PB, Mahan TE, Landreth GE, Castellano JM, Jiang H, Cirrito JR, Holtzman DM. In vivo measurement of apolipoprotein E from the brain interstitial fluid using microdialysis. Mol Neurodegener. 2013;8:13.CrossRefPubMedPubMedCentral Ulrich JD, Burchett JM, Restivo JL, Schuler DR, Verghese PB, Mahan TE, Landreth GE, Castellano JM, Jiang H, Cirrito JR, Holtzman DM. In vivo measurement of apolipoprotein E from the brain interstitial fluid using microdialysis. Mol Neurodegener. 2013;8:13.CrossRefPubMedPubMedCentral
41.
42.
Zurück zum Zitat Chantepie S, Bochem AE, Chapman MJ, Hovingh GK, Kontush A. High-density lipoprotein (HDL) particle subpopulations in heterozygous cholesteryl ester transfer protein (CETP) deficiency: maintenance of antioxidative activity. PLoS One. 2012;7:e49336.CrossRefPubMedPubMedCentral Chantepie S, Bochem AE, Chapman MJ, Hovingh GK, Kontush A. High-density lipoprotein (HDL) particle subpopulations in heterozygous cholesteryl ester transfer protein (CETP) deficiency: maintenance of antioxidative activity. PLoS One. 2012;7:e49336.CrossRefPubMedPubMedCentral
43.
Zurück zum Zitat Miida T, Yamazaki F, Sakurai M, Wada R, Yamadera T, Asami K, Hoshiyama M, Tanaka A, Inano K, Okada M. The apolipoprotein E content of HDL in cerebrospinal fluid is higher in children than in adults. Clin Chem. 1999;45:1294–6.PubMed Miida T, Yamazaki F, Sakurai M, Wada R, Yamadera T, Asami K, Hoshiyama M, Tanaka A, Inano K, Okada M. The apolipoprotein E content of HDL in cerebrospinal fluid is higher in children than in adults. Clin Chem. 1999;45:1294–6.PubMed
44.
Zurück zum Zitat Weisgraber KH. Apolipoprotein E: structure-function relationships. Adv Protein Chem. 1994;45:249–302.CrossRefPubMed Weisgraber KH. Apolipoprotein E: structure-function relationships. Adv Protein Chem. 1994;45:249–302.CrossRefPubMed
45.
Zurück zum Zitat Nayyar G, Garber DW, Palgunachari MN, Monroe CE, Keenum TD, Handattu SP, Mishra VK, Anantharamaiah GM. Apolipoprotein E mimetic is more effective than apolipoprotein A-I mimetic in reducing lesion formation in older female apo E null mice. Atherosclerosis. 2012;224:326–31.CrossRefPubMedPubMedCentral Nayyar G, Garber DW, Palgunachari MN, Monroe CE, Keenum TD, Handattu SP, Mishra VK, Anantharamaiah GM. Apolipoprotein E mimetic is more effective than apolipoprotein A-I mimetic in reducing lesion formation in older female apo E null mice. Atherosclerosis. 2012;224:326–31.CrossRefPubMedPubMedCentral
46.
48.
Zurück zum Zitat Marban C, Forouzanfar F, Ait-Ammar A, Fahmi F, El Mekdad H, Daouad F, Rohr O, Schwartz C. Targeting the brain reservoirs: toward an HIV cure. Front Immunol. 2016;7:397.CrossRefPubMedPubMedCentral Marban C, Forouzanfar F, Ait-Ammar A, Fahmi F, El Mekdad H, Daouad F, Rohr O, Schwartz C. Targeting the brain reservoirs: toward an HIV cure. Front Immunol. 2016;7:397.CrossRefPubMedPubMedCentral
49.
Zurück zum Zitat Gray LR, Roche M, Flynn JK, Wesselingh SL, Gorry PR, Churchill MJ. Is the central nervous system a reservoir of HIV-1? Curr Opin HIV AIDS. 2014;9:552–8.CrossRefPubMedPubMedCentral Gray LR, Roche M, Flynn JK, Wesselingh SL, Gorry PR, Churchill MJ. Is the central nervous system a reservoir of HIV-1? Curr Opin HIV AIDS. 2014;9:552–8.CrossRefPubMedPubMedCentral
50.
Zurück zum Zitat Churchill MJ, Gorry PR, Cowley D, Lal L, Sonza S, Purcell DF, Thompson KA, Gabuzda D, McArthur JC, Pardo CA, Wesselingh SL. Use of laser capture microdissection to detect integrated HIV-1 DNA in macrophages and astrocytes from autopsy brain tissues. J Neuro-Oncol. 2006;12:146–52. Churchill MJ, Gorry PR, Cowley D, Lal L, Sonza S, Purcell DF, Thompson KA, Gabuzda D, McArthur JC, Pardo CA, Wesselingh SL. Use of laser capture microdissection to detect integrated HIV-1 DNA in macrophages and astrocytes from autopsy brain tissues. J Neuro-Oncol. 2006;12:146–52.
51.
Zurück zum Zitat Thompson KA, Cherry CL, Bell JE, McLean CA. Brain cell reservoirs of latent virus in presymptomatic HIV-infected individuals. Am J Pathol. 2011;179:1623–9.CrossRefPubMedPubMedCentral Thompson KA, Cherry CL, Bell JE, McLean CA. Brain cell reservoirs of latent virus in presymptomatic HIV-infected individuals. Am J Pathol. 2011;179:1623–9.CrossRefPubMedPubMedCentral
52.
Zurück zum Zitat Watkins BA, Dorn HH, Kelly WB, Armstrong RC, Potts BJ, Michaels F, Kufta CV, Dubois-Dalcq M. Specific tropism of HIV-1 for microglial cells in primary human brain cultures. Science. 1990;249:549–53.CrossRefPubMed Watkins BA, Dorn HH, Kelly WB, Armstrong RC, Potts BJ, Michaels F, Kufta CV, Dubois-Dalcq M. Specific tropism of HIV-1 for microglial cells in primary human brain cultures. Science. 1990;249:549–53.CrossRefPubMed
53.
Zurück zum Zitat Wiley CA, Schrier RD, Nelson JA, Lampert PW, Oldstone MB. Cellular localization of human immunodeficiency virus infection within the brains of acquired immune deficiency syndrome patients. Proc Natl Acad Sci U S A. 1986;83:7089–93.CrossRefPubMedPubMedCentral Wiley CA, Schrier RD, Nelson JA, Lampert PW, Oldstone MB. Cellular localization of human immunodeficiency virus infection within the brains of acquired immune deficiency syndrome patients. Proc Natl Acad Sci U S A. 1986;83:7089–93.CrossRefPubMedPubMedCentral
54.
Zurück zum Zitat Chauhan A, Mehla R, Vijayakumar TS, Handy I. Endocytosis-mediated HIV-1 entry and its significance in the elusive behavior of the virus in astrocytes. Virology. 2014;456-457:1–19.CrossRefPubMed Chauhan A, Mehla R, Vijayakumar TS, Handy I. Endocytosis-mediated HIV-1 entry and its significance in the elusive behavior of the virus in astrocytes. Virology. 2014;456-457:1–19.CrossRefPubMed
55.
Zurück zum Zitat Carter GC, Bernstone L, Baskaran D, James W. HIV-1 infects macrophages by exploiting an endocytic route dependent on dynamin, Rac1 and Pak1. Virology. 2011;409:234–50.CrossRefPubMed Carter GC, Bernstone L, Baskaran D, James W. HIV-1 infects macrophages by exploiting an endocytic route dependent on dynamin, Rac1 and Pak1. Virology. 2011;409:234–50.CrossRefPubMed
56.
Zurück zum Zitat Trillo-Pazos G, Diamanturos A, Rislove L, Menza T, Chao W, Belem P, Sadiq S, Morgello S, Sharer L, Volsky DJ. Detection of HIV-1 DNA in microglia/macrophages, astrocytes and neurons isolated from brain tissue with HIV-1 encephalitis by laser capture microdissection. Brain Pathol. 2003;13:144–54.CrossRefPubMed Trillo-Pazos G, Diamanturos A, Rislove L, Menza T, Chao W, Belem P, Sadiq S, Morgello S, Sharer L, Volsky DJ. Detection of HIV-1 DNA in microglia/macrophages, astrocytes and neurons isolated from brain tissue with HIV-1 encephalitis by laser capture microdissection. Brain Pathol. 2003;13:144–54.CrossRefPubMed
57.
Zurück zum Zitat Churchill MJ, Wesselingh SL, Cowley D, Pardo CA, McArthur JC, Brew BJ, Gorry PR. Extensive astrocyte infection is prominent in human immunodeficiency virus-associated dementia. Ann Neurol. 2009;66:253–8.CrossRefPubMed Churchill MJ, Wesselingh SL, Cowley D, Pardo CA, McArthur JC, Brew BJ, Gorry PR. Extensive astrocyte infection is prominent in human immunodeficiency virus-associated dementia. Ann Neurol. 2009;66:253–8.CrossRefPubMed
58.
Zurück zum Zitat Johnson TP, Patel K, Johnson KR, Maric D, Calabresi PA, Hasbun R, Nath A. Induction of IL-17 and nonclassical T-cell activation by HIV-Tat protein. Proc Natl Acad Sci U S A. 2013;110:13588–93.CrossRefPubMedPubMedCentral Johnson TP, Patel K, Johnson KR, Maric D, Calabresi PA, Hasbun R, Nath A. Induction of IL-17 and nonclassical T-cell activation by HIV-Tat protein. Proc Natl Acad Sci U S A. 2013;110:13588–93.CrossRefPubMedPubMedCentral
59.
Zurück zum Zitat Tyagi M, Rusnati M, Presta M, Giacca M. Internalization of HIV-1 tat requires cell surface heparan sulfate proteoglycans. J Biol Chem. 2001;276:3254–61.CrossRefPubMed Tyagi M, Rusnati M, Presta M, Giacca M. Internalization of HIV-1 tat requires cell surface heparan sulfate proteoglycans. J Biol Chem. 2001;276:3254–61.CrossRefPubMed
60.
Zurück zum Zitat Vives E. Cellular uptake [correction of utake] of the Tat peptide: an endocytosis mechanism following ionic interactions. J Mol Recognit. 2003;16:265–71.CrossRefPubMed Vives E. Cellular uptake [correction of utake] of the Tat peptide: an endocytosis mechanism following ionic interactions. J Mol Recognit. 2003;16:265–71.CrossRefPubMed
61.
Zurück zum Zitat Ensoli B, Buonaguro L, Barillari G, Fiorelli V, Gendelman R, Morgan RA, Wingfield P, Gallo RC. Release, uptake, and effects of extracellular human immunodeficiency virus type 1 Tat protein on cell growth and viral transactivation. J Virol. 1993;67:277–87.PubMedPubMedCentral Ensoli B, Buonaguro L, Barillari G, Fiorelli V, Gendelman R, Morgan RA, Wingfield P, Gallo RC. Release, uptake, and effects of extracellular human immunodeficiency virus type 1 Tat protein on cell growth and viral transactivation. J Virol. 1993;67:277–87.PubMedPubMedCentral
62.
Zurück zum Zitat Westendorp MO, Frank R, Ochsenbauer C, Stricker K, Dhein J, Walczak H, Debatin KM, Krammer PH. Sensitization of T cells to CD95-mediated apoptosis by HIV-1 Tat and gp120. Nature. 1995;375:497–500.CrossRefPubMed Westendorp MO, Frank R, Ochsenbauer C, Stricker K, Dhein J, Walczak H, Debatin KM, Krammer PH. Sensitization of T cells to CD95-mediated apoptosis by HIV-1 Tat and gp120. Nature. 1995;375:497–500.CrossRefPubMed
63.
Zurück zum Zitat Xiao H, Neuveut C, Tiffany HL, Benkirane M, Rich EA, Murphy PM, Jeang KT. Selective CXCR4 antagonism by Tat: implications for in vivo expansion of coreceptor use by HIV-1. Proc Natl Acad Sci U S A. 2000;97:11466–71.CrossRefPubMedPubMedCentral Xiao H, Neuveut C, Tiffany HL, Benkirane M, Rich EA, Murphy PM, Jeang KT. Selective CXCR4 antagonism by Tat: implications for in vivo expansion of coreceptor use by HIV-1. Proc Natl Acad Sci U S A. 2000;97:11466–71.CrossRefPubMedPubMedCentral
64.
Zurück zum Zitat Ensoli B, Gendelman R, Markham P, Fiorelli V, Colombini S, Raffeld M, Cafaro A, Chang HK, Brady JN, Gallo RC. Synergy between basic fibroblast growth factor and HIV-1 Tat protein in induction of Kaposi’s sarcoma. Nature. 1994;371:674–80.CrossRefPubMed Ensoli B, Gendelman R, Markham P, Fiorelli V, Colombini S, Raffeld M, Cafaro A, Chang HK, Brady JN, Gallo RC. Synergy between basic fibroblast growth factor and HIV-1 Tat protein in induction of Kaposi’s sarcoma. Nature. 1994;371:674–80.CrossRefPubMed
65.
Zurück zum Zitat Frankel AD, Bredt DS, Pabo CO. Tat protein from human immunodeficiency virus forms a metal-linked dimer. Science. 1988;240:70–3.CrossRefPubMed Frankel AD, Bredt DS, Pabo CO. Tat protein from human immunodeficiency virus forms a metal-linked dimer. Science. 1988;240:70–3.CrossRefPubMed
66.
Zurück zum Zitat Magnuson DS, Knudsen BE, Geiger JD, Brownstone RM, Nath A. Human immunodeficiency virus type 1 tat activates non-N-methyl-D-aspartate excitatory amino acid receptors and causes neurotoxicity. Ann Neurol. 1995;37:373–80.CrossRefPubMed Magnuson DS, Knudsen BE, Geiger JD, Brownstone RM, Nath A. Human immunodeficiency virus type 1 tat activates non-N-methyl-D-aspartate excitatory amino acid receptors and causes neurotoxicity. Ann Neurol. 1995;37:373–80.CrossRefPubMed
67.
Zurück zum Zitat Mediouni S, Darque A, Baillat G, Ravaux I, Dhiver C, Tissot-Dupont H, Mokhtari M, Moreau H, Tamalet C, Brunet C, et al. Antiretroviral therapy does not block the secretion of the human immunodeficiency virus tat protein. Infect Disord Drug Targets. 2012;12:81–6.CrossRefPubMed Mediouni S, Darque A, Baillat G, Ravaux I, Dhiver C, Tissot-Dupont H, Mokhtari M, Moreau H, Tamalet C, Brunet C, et al. Antiretroviral therapy does not block the secretion of the human immunodeficiency virus tat protein. Infect Disord Drug Targets. 2012;12:81–6.CrossRefPubMed
68.
Zurück zum Zitat Mediouni S, Watkins JD, Pierres M, Bole A, Loret EP, Baillat G. A monoclonal antibody directed against a conformational epitope of the HIV-1 trans-activator (Tat) protein neutralizes cross-clade. J Biol Chem. 2012;287:11942–50.CrossRefPubMedPubMedCentral Mediouni S, Watkins JD, Pierres M, Bole A, Loret EP, Baillat G. A monoclonal antibody directed against a conformational epitope of the HIV-1 trans-activator (Tat) protein neutralizes cross-clade. J Biol Chem. 2012;287:11942–50.CrossRefPubMedPubMedCentral
69.
Zurück zum Zitat Hudson L, Liu J, Nath A, Jones M, Raghavan R, Narayan O, Male D, Everall I. Detection of the human immunodeficiency virus regulatory protein tat in CNS tissues. J Neuro-Oncol. 2000;6:145–55. Hudson L, Liu J, Nath A, Jones M, Raghavan R, Narayan O, Male D, Everall I. Detection of the human immunodeficiency virus regulatory protein tat in CNS tissues. J Neuro-Oncol. 2000;6:145–55.
70.
Zurück zum Zitat Pomerantz RJ, Trono D, Feinberg MB, Baltimore D. Cells nonproductively infected with HIV-1 exhibit an aberrant pattern of viral RNA expression: a molecular model for latency. Cell. 1990;61:1271–6.CrossRefPubMed Pomerantz RJ, Trono D, Feinberg MB, Baltimore D. Cells nonproductively infected with HIV-1 exhibit an aberrant pattern of viral RNA expression: a molecular model for latency. Cell. 1990;61:1271–6.CrossRefPubMed
71.
Zurück zum Zitat Tornatore C, Meyers K, Atwood W, Conant K, Major E. Temporal patterns of human immunodeficiency virus type 1 transcripts in human fetal astrocytes. J Virol. 1994;68:93–102.PubMedPubMedCentral Tornatore C, Meyers K, Atwood W, Conant K, Major E. Temporal patterns of human immunodeficiency virus type 1 transcripts in human fetal astrocytes. J Virol. 1994;68:93–102.PubMedPubMedCentral
72.
Zurück zum Zitat Sykova E. Extracellular space volume and geometry of the rat brain after ischemia and central injury. Adv Neurol. 1997;73:121–35.PubMed Sykova E. Extracellular space volume and geometry of the rat brain after ischemia and central injury. Adv Neurol. 1997;73:121–35.PubMed
73.
Zurück zum Zitat Cardarelli F, Digiacomo L, Marchini C, Amici A, Salomone F, Fiume G, Rossetta A, Gratton E, Pozzi D, Caracciolo G. The intracellular trafficking mechanism of Lipofectamine-based transfection reagents and its implication for gene delivery. Sci Rep. 2016;6:25879.CrossRefPubMedPubMedCentral Cardarelli F, Digiacomo L, Marchini C, Amici A, Salomone F, Fiume G, Rossetta A, Gratton E, Pozzi D, Caracciolo G. The intracellular trafficking mechanism of Lipofectamine-based transfection reagents and its implication for gene delivery. Sci Rep. 2016;6:25879.CrossRefPubMedPubMedCentral
74.
Zurück zum Zitat Lonn P, Kacsinta AD, Cui XS, Hamil AS, Kaulich M, Gogoi K, Dowdy SF. Enhancing endosomal escape for intracellular delivery of macromolecular biologic therapeutics. Sci Rep. 2016;6:32301.CrossRefPubMedPubMedCentral Lonn P, Kacsinta AD, Cui XS, Hamil AS, Kaulich M, Gogoi K, Dowdy SF. Enhancing endosomal escape for intracellular delivery of macromolecular biologic therapeutics. Sci Rep. 2016;6:32301.CrossRefPubMedPubMedCentral
75.
Zurück zum Zitat Herz J, Kowal RC, Goldstein JL, Brown MS. Proteolytic processing of the 600 kd low density lipoprotein receptor-related protein (LRP) occurs in a trans-Golgi compartment. EMBO J. 1990;9:1769–76.PubMedPubMedCentral Herz J, Kowal RC, Goldstein JL, Brown MS. Proteolytic processing of the 600 kd low density lipoprotein receptor-related protein (LRP) occurs in a trans-Golgi compartment. EMBO J. 1990;9:1769–76.PubMedPubMedCentral
76.
Zurück zum Zitat Neels JG, van Den Berg BM, Lookene A, Olivecrona G, Pannekoek H, van Zonneveld AJ. The second and fourth cluster of class A cysteine-rich repeats of the low density lipoprotein receptor-related protein share ligand-binding properties. J Biol Chem. 1999;274:31305–11.CrossRefPubMed Neels JG, van Den Berg BM, Lookene A, Olivecrona G, Pannekoek H, van Zonneveld AJ. The second and fourth cluster of class A cysteine-rich repeats of the low density lipoprotein receptor-related protein share ligand-binding properties. J Biol Chem. 1999;274:31305–11.CrossRefPubMed
77.
Zurück zum Zitat Laatsch A, Panteli M, Sornsakrin M, Hoffzimmer B, Grewal T, Heeren J. Low density lipoprotein receptor-related protein 1 dependent endosomal trapping and recycling of apolipoprotein E. PLoS One. 2012;7:e29385.CrossRefPubMedPubMedCentral Laatsch A, Panteli M, Sornsakrin M, Hoffzimmer B, Grewal T, Heeren J. Low density lipoprotein receptor-related protein 1 dependent endosomal trapping and recycling of apolipoprotein E. PLoS One. 2012;7:e29385.CrossRefPubMedPubMedCentral
78.
Zurück zum Zitat Cruchaga C, Kauwe JS, Nowotny P, Bales K, Pickering EH, Mayo K, Bertelsen S, Hinrichs A, Alzheimer’s Disease Neuroimaging I, Fagan AM, et al. Cerebrospinal fluid APOE levels: an endophenotype for genetic studies for Alzheimer’s disease. Hum Mol Genet. 2012;21:4558–71.CrossRefPubMedPubMedCentral Cruchaga C, Kauwe JS, Nowotny P, Bales K, Pickering EH, Mayo K, Bertelsen S, Hinrichs A, Alzheimer’s Disease Neuroimaging I, Fagan AM, et al. Cerebrospinal fluid APOE levels: an endophenotype for genetic studies for Alzheimer’s disease. Hum Mol Genet. 2012;21:4558–71.CrossRefPubMedPubMedCentral
79.
Zurück zum Zitat Ruiz J, Kouiavskaia D, Migliorini M, Robinson S, Saenko EL, Gorlatova N, Li D, Lawrence D, Hyman BT, Weisgraber KH, Strickland DK. The apoE isoform binding properties of the VLDL receptor reveal marked differences from LRP and the LDL receptor. J Lipid Res. 2005;46:1721–31.CrossRefPubMed Ruiz J, Kouiavskaia D, Migliorini M, Robinson S, Saenko EL, Gorlatova N, Li D, Lawrence D, Hyman BT, Weisgraber KH, Strickland DK. The apoE isoform binding properties of the VLDL receptor reveal marked differences from LRP and the LDL receptor. J Lipid Res. 2005;46:1721–31.CrossRefPubMed
80.
Zurück zum Zitat Narita M, Holtzman DM, Fagan AM, LaDu MJ, Yu L, Han X, Gross RW, Bu G, Schwartz AL. Cellular catabolism of lipid poor apolipoprotein E via cell surface LDL receptor-related protein. J Biochem. 2002;132:743–9.CrossRefPubMed Narita M, Holtzman DM, Fagan AM, LaDu MJ, Yu L, Han X, Gross RW, Bu G, Schwartz AL. Cellular catabolism of lipid poor apolipoprotein E via cell surface LDL receptor-related protein. J Biochem. 2002;132:743–9.CrossRefPubMed
81.
Zurück zum Zitat Croy JE, Brandon T, Komives EA. Two apolipoprotein E mimetic peptides, ApoE(130-149) and ApoE(141-155)2, bind to LRP1. Biochemistry. 2004;43:7328–35.CrossRefPubMed Croy JE, Brandon T, Komives EA. Two apolipoprotein E mimetic peptides, ApoE(130-149) and ApoE(141-155)2, bind to LRP1. Biochemistry. 2004;43:7328–35.CrossRefPubMed
82.
Zurück zum Zitat Bell RD, Winkler EA, Singh I, Sagare AP, Deane R, Wu Z, Holtzman DM, Betsholtz C, Armulik A, Sallstrom J, et al. Apolipoprotein E controls cerebrovascular integrity via cyclophilin A. Nature. 2012;485:512–6.CrossRefPubMedPubMedCentral Bell RD, Winkler EA, Singh I, Sagare AP, Deane R, Wu Z, Holtzman DM, Betsholtz C, Armulik A, Sallstrom J, et al. Apolipoprotein E controls cerebrovascular integrity via cyclophilin A. Nature. 2012;485:512–6.CrossRefPubMedPubMedCentral
83.
Zurück zum Zitat Hatters DM, Peters-Libeu CA, Weisgraber KH. Apolipoprotein E structure: insights into function. Trends Biochem Sci. 2006;31:445–54.CrossRefPubMed Hatters DM, Peters-Libeu CA, Weisgraber KH. Apolipoprotein E structure: insights into function. Trends Biochem Sci. 2006;31:445–54.CrossRefPubMed
84.
Zurück zum Zitat Nguyen D, Dhanasekaran P, Phillips MC, Lund-Katz S. Molecular mechanism of apolipoprotein E binding to lipoprotein particles. Biochemistry. 2009;48:3025–32.CrossRefPubMedPubMedCentral Nguyen D, Dhanasekaran P, Phillips MC, Lund-Katz S. Molecular mechanism of apolipoprotein E binding to lipoprotein particles. Biochemistry. 2009;48:3025–32.CrossRefPubMedPubMedCentral
Metadaten
Titel
Apolipoprotein E isoform dependently affects Tat-mediated HIV-1 LTR transactivation
verfasst von
Nabab Khan
Gaurav Datta
Jonathan D. Geiger
Xuesong Chen
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
Journal of Neuroinflammation / Ausgabe 1/2018
Elektronische ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-018-1129-1

Weitere Artikel der Ausgabe 1/2018

Journal of Neuroinflammation 1/2018 Zur Ausgabe

Neu in den Fachgebieten Neurologie und Psychiatrie