Skip to main content

Peroxisome proliferator-activated receptor-gamma: potential molecular therapeutic target for HIV-1-associated brain inflammation

Abstract

Background

Despite the use of combination antiretroviral therapy for the treatment of HIV-1 infection, cognitive impairments remain prevalent due to persistent viral replication and associated brain inflammation. Primary cellular targets of HIV-1 in the brain are macrophages, microglia, and to a certain extent astrocytes which in response to infection release inflammatory markers, viral proteins [i.e., glycoprotein 120 (gp120)] and exhibit impaired glutamate uptake. Peroxisome proliferator-activated receptors (PPARs) are members of the nuclear receptor superfamily of ligand-activated transcription factors. Compelling evidence suggests that PPARĪ³ exerts anti-inflammatory properties in neurological disorders. The goal of this study was to examine the role of PPARĪ³ in the context of HIV-1ADA gp120-induced inflammation in vitro, in primary cultures of rat astrocytes and microglia, and in vivo, in a rodent model of HIV-1ADA gp120-associated brain inflammation.

Methods

Primary mixed cultures of rat astrocytes and microglia were treated with PPARĪ³ agonists (rosiglitazone or pioglitazone) and exposed to HIV-1ADA gp120. Inflammatory cytokines and indicator of oxidative stress response (TNFĪ±, IL-1Ī², iNOS) were measured using qPCR, and glutamate transporter (GLT-1) was quantified by immunoblotting. In vivo, rats were administered an intracerebroventricular injection of HIV-1ADA gp120 and an intraperitoneal injection of PPARĪ³ agonist (rosiglitazone) or co-administration with PPARĪ³ antagonist (GW9662). qPCR and immunoblotting analyses were applied to measure inflammatory markers, GLT-1 and PPARĪ³.

Results

In primary mixed cultures of rat astrocytes and microglia, HIV-1ADA gp120 exposure resulted in a significant elevation of inflammatory markers and a decrease in GLT-1 expression which were significantly attenuated with rosiglitazone or pioglitazone treatment. Similarly, in vivo, treatment with rosiglitazone reversed the gp120-mediated inflammatory response and downregulation of GLT-1. Furthermore, we demonstrated that the anti-inflammatory effects of PPARĪ³ agonist rosiglitazone were mediated through inhibition of NF-ĪŗB.

Conclusion

Our data demonstrate that gp120 can induce an inflammatory response and decrease expression of GLT-1 in the brain in vitro and in vivo. We have also successfully shown that these effects can be reversed by treatment with PPARĪ³ agonists, rosiglitazone or pioglitazone. Together our data suggest that targeting PPARĪ³ signaling may provide an option for preventing/treating HIV-associated brain inflammation.

Background

The entry of the human immunodeficiency virus (HIV-1) into the central nervous system (CNS) occurs early in the course of infection either as a cell-free virion or encased within infected macrophages [1]. Some reports also document that HIV-1 can cross the blood-brain barrier (BBB) through a receptor-mediated transcytosis possibly using the mannose-6-receptor [2]. In the CNS, the major targets of HIV-1 are mononuclear phagocytes (e.g., perivascular macrophages and brain resident microglial cells) and to lesser degree astrocytes. In response to HIV-1, microglia and astrocytes become activated and secrete pro-inflammatory cytokines [i.e., tumor necrosis factor-Ī± (TNFĪ±), interleukin-1Ī² (IL-1Ī²), interleukin-6 (IL-6), interleukin-8 (IL-8)] and neurotoxins [i.e., arachidonic/quinolinic acid and metabolites, platelet-activating factor, neurotoxic amines, reactive oxygen species (ROS), nitric oxide (NO), and glutamate] [3]. Although neurons do not appear to be directly infected by HIV-1, the prolonged exposure to inflammatory, neurotoxic, and oxidative stress markers during infection can cause neuronal injury and death [4]. HIV-1 viral proteins such as envelope glycoprotein (gp120), transactivator of transcription (Tat), and viral protein R (Vpr) are also known to be neurotoxic upon release from infected cells [3, 5]. It has been postulated that mechanisms triggering neuronal apoptosis involve viral protein interactions with neuronal chemokine receptors, excitotoxicity due to glutamate accumulation, caspase activation, loss of mitochondrial membrane potential, and DNA fragmentation [3]. We have previously demonstrated that R5 tropic HIV-1ADA gp120 can mediate secretion of pro-inflammatory cytokines and oxidative stress markers by interacting with CCR5 chemokine receptor in primary cultures of human and rodent astrocytes, as well as in an in vivo rodent model of gp120-associated brain inflammation [6,7,8].

Despite receiving highly active antiretroviral therapy (HAART), up to 50% of infected individuals canĀ develop HIV-1-associated neurocognitive disorders (HAND) which include memory, motor, and behavioral deficits, and can affect quality of life and mortality rate in these patients [9, 10]. The underlying mechanism for HAND remains poorly understood; however, a contributing factor may be chronic brain inflammation due to low level of HIV-1 replication in viral reservoirs such as microglia, and secretion or shedding of viral proteins (e.g., gp120, Vpr). Other contributing factors include age, low CD4+ T-cell nadir count, and comorbidities [9]. Currently, there are no effective treatments for HAND, and although HAART significantly prolongs lives of HIV-1-infected patients, variable effects have been reported on neurocognitive performance, and in some cases, certain antiretroviral drugs (ARVs) have been associated with neurotoxicity [11]. ARVs which are available for clinical use allow for systemic suppression of peripheral viral load; however, treating HIV in the brain remains a challenge partly due to the fact that several ARVs exhibit poor permeability across the BBB and into glial cells. In particular, protease inhibitors and nucleoside reverse transcriptase inhibitors display low brain penetration and do not reach therapeutic concentrations within the CNS, potentially allowing the brain to become a sanctuary for HIV-1 [12, 13]. Insufficient ARV concentrations in the brain could permit continuous HIV-1 replication and subsequent emergence of drug resistance viral strains despite acceptable control of the virus in the periphery [14]. In addition to low brain permeability, it is also important to note that the majority of ARVs do not exhibit direct anti-inflammatory properties. Therefore, identifying alternative therapeutic approaches that prevent release of neurotoxic factors from glial cells is critical for the treatment of HIV-associated brain inflammation and neurological disorders.

A variety of potential biomarkers have been identified in association with HAND. Several studies have shown, in HIV-infected individuals who develop HAND, markers of immune activation (neopterin, sCD14), cytokine expression (TNFĪ±), and oxidative stress are more pronounced than in HIV individuals without cognitive impairments [15]. Abnormal glutamate homeostasis has also been observed in HIV-1-infected patients, where an increase in glutamate is observed in the cerebrospinal fluid (CSF) of patients with HAND as compared to healthy controls [16]. Furthermore, HIV-1 viral proteins Tat and gp120 have shown to decrease glial and synaptic uptake of glutamate [17,18,19].

With the increased prevalence of HAND among HIV-1-infected individuals, and the lack of effective therapy, it is critical to identify potential targets for the treatment of HAND [9]. In the past decade, there has been growing interest in the peroxisome proliferator-activated receptors (PPARs) ligand-activated transcription factors belonging to the nuclear receptors for steroid, thyroid hormones, and retinoids. These receptors play major roles in lipid homeostasis and glucose regulation [20]. Additionally, PPAR agonists can exhibit anti-inflammatory and antioxidant effects in several models of CNS disorders such as ischemic stroke and Alzheimerā€™s and Parkinsonā€™s diseases [21,22,23]. Several studies have used both in vitro and in vivo models to demonstrate PPAR-mediated attenuation of the release of pro-inflammatory cytokines and oxidative stress markers [24]. In the context of HIV-1, there is also evidence suggesting that PPARĪ³ and to a lesser extent PPARĪ± agonists can play a neuroprotective role [5, 25, 26]. It has also been demonstrated that PPARĪ³ agonist rosiglitazone can exhibit direct anti-HIV effects in different cell types such as Th1Th17 cells and monocyte-derived macrophages [27]; therefore, this isoform is of further interest. The protective anti-inflammatory effects of PPARĪ³ have been shown to be partly mediated through transrepression of the redox regulated transcription factor nuclear factor kappa B (NF-ĪŗB) [28, 29].

The goal of this project was to investigate the role of PPARĪ³: (i) in suppression of inflammation and glutamate transporter 1 (GLT-1) dysregulation, in vitro, in primary cultures of rat microglia and astrocytes exposed to HIV-1ADA gp120, and (ii) in vivo, in a rat model of HIV-1 associated brain inflammation.

Methods

Materials

HIV-1ADA gp120 full-length recombinant protein (Clade B; R5-tropic) was obtained from immunodiagnostics Inc. (Woburn, Massachusetts, USA). PPARĪ³ agonists rosiglitazone and pioglitazone and PPARĪ³ antagonist 2-chloro-5-nitro-N-phenylbenzamide (GW9662) were purchased from Cayman Chemicals (Ann Arbor, Michigan, USA). Rabbit polyclonal anti-PPARĪ³ (ab-6643), mouse monoclonal anti-CD11b/c (ab-1211), and rabbit polyclonal anti-EAAT2 (ab-41621) antibodies were purchased from Abcam Inc. (Boston, MA, USA). Rabbit polyclonal anti p-NF-ĪŗB p65Ser536 (sc-33020) and mouse monoclonal Ī²ā€“Actin (sc-47778) antibodies were obtained from Santa Cruz Biotechnology (Dallas, Texas, USA). Rabbit polyclonal antibody against glial fibrillary acidic protein (GFAP) and horse radish peroxidase (HRP) conjugated secondary antibodies (anti-mouse and anti-rabbit) were obtained from Sigma Aldrich (Missisauga, ON, Canada). Alexa fluor 488 and 594 (anti-rabbit or anti-mouse), 4ā€²,6-diamidino-2-phenylindole hydrochloride (DAPI), western blot stripping solution, enhanced chemiluminescent reagents, and TRIzol were purchased from ThermoFisher Scientific (Waltham, MA, USA). High capacity reverse transcriptase cDNA synthesis kit and TaqMan FastMix were obtained from Applied Biosystems (Foster City, CA, USA) and Quanta Biosciences Inc. (Gaithersburg, Maryland, USA), respectively. Mixed astrocyte-microglia culture medium was prepared from minimum essential medium (OCI MEM H17 without antibiotic), gentamicin (Cat# 15750-060), Horse Serum (Cat# 16050-122) and Fetal Bovine Serum (Cat# 26140-079) from ThermoFisher Scientific (Waltham, MA, USA). 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) reagent was purchased from Sigma Aldrich (Mississauga, ON, Canada).

Cell cultures

Primary cultures of rat astrocytes and microglia were prepared as described previously in our laboratory [6] with a few modifications. All procedures were carried out in accordance with the University of Toronto Animal Care Committee and the Province of Ontario Animals for Research Act. In brief, whole brain isolates from 1- to 3-day-old neonatal Wistar rats (Charles River Laboratories, St. Constant, QC, Canada) were collected by cervical dislocation. Cerebral cortices were dissected and subjected to enzymatic digestion for 30Ā min in serum-free medium containing 2.0Ā mg/mL porcine pancreatic trypsin Sigma Aldrich (Mississauga, ON, Canada) and 0.005% DNase I purchased from Roche, Applied Science (Laval, QC, Canada). The tissue was then mechanically disaggregated using a cell dissociation kit from Sigma-Aldrich (Mississauga, ON, Canada) to yield a mixed glial cell suspension. The cell suspension was then centrifuged for 10Ā min at 100Ā g and resuspended in primary glial culture medium, which consisted of minimum essential medium supplemented with 5% horse serum, 5% fetal bovine serum, and 5Ā Ī¼g/ml gentamicin. The cells were plated onto 25-cm2 polystyrene tissue culture flasks (Sarstedt, St. Leonard, PQ, Canada) and incubated in fresh medium at 37Ā Ā°C, in 5% CO2/95% air for 7 to 10Ā days until confluence was attained. For pure astrocyte cultures, the cells were then placed on an orbital shaker at 120Ā rpm for 6Ā h to remove microglia. Cells in culture were characterized for their purity and were assessed by morphological analysis and immunostaining for standard biochemical markers (e.g., glial fibrillary acidic protein for astrocytes, cd11b/c for microglia) (Fig.Ā 1). K-562, the chronic myelogenous leukemia human cell lysate, was purchased from Santa Cruz Biotechnology (Dallas, Texas, USA) and was used as a positive control for p-NF-ĪŗB p65Ser536 in immunoblotting experiments. HepG2, hepatocyte carcinoma cell line, was purchased from ATCC (Manassas, Virginia, USA); cell lysates were prepared in our laboratory and used as a positive control for PPARĪ³.

Fig. 1
figure 1

Immunocytochemical analysis of primary cultures of mixed rat astrocytes and microglia. Cells were immunostained with a DAPI, nuclear marker; b GFAP (1:200, dilution) astrocyte marker; (c) CD11b/c (1:20, dilution) microglial marker; d merged image. Cells were visualized using a confocal microscope (LSM 700, Carl Zeiss) operated with ZEN software using a 40Ɨ objective lens. Scale bar, 75Ā Ī¼M

Immunocytochemical analysis

Immunofluorescence experiments were performed as previously described in our laboratory [30] with a few modifications. In brief, cell monolayers grown on glass coverslips were fixed with 4% paraformaldehyde (PFA) for 20Ā min at room temperature. After fixation, cells were washed in PBS and permeabilized with 0.1% Triton X-100 for 5Ā min at room temperature. Fixed cells were blocked with 0.1% (m/v) bovine serum albumin and 0.1% (m/v) skim milk in PBS for 1Ā h before primary antibody incubation for 1.5Ā h at room temperature or overnight at 4Ā Ā°C. The rabbit polyclonal (anti-GFAP, 1:200 dilution) and mouse monoclonal (anti-CD11b/c, 1:20) antibodies were used for markers of astrocytes and microglia, respectively. After primary antibody incubation, cells were washed with PBS by gentle agitation and followed by incubation with anti-mouse Alexa Fluor 594 or anti-rabbit Alexa Fluor 488 conjugated secondary antibody (both in 1:500 dilution). Staining in the absence of primary antibodies was used as a negative control. After secondary antibody incubation, cells were washed again with PBS and mounted on a 76Ā Ć—Ā 26Ā mm microscope slide (VWR, West Chester, PA) using VECTASHIELD mounting solution containing DAPI. Cells were visualized using a confocal microscope (LSM 700, Carl Zeiss) operated with ZEN software.

HIV-1ADA gp120 cell treatment

All treatments were performed on monolayers of primary cultures of either mixed rat astrocytes and microglia or pure astrocytes grown in 25-cm2 tissue culture flasks. At the beginning of each experiment, culture medium was aspirated and fresh medium containing 5% fetal bovine serum and 5% horse serum with 5Ā nM HIVADA gp120 was added. In our pilot experiments, using less than 5Ā nM of gp120 produced more variable inflammatory responses and concentrations higher than 5Ā nM did not induce higher responses. Furthermore, in our present study as well as other published reports from our laboratory [6, 7], we have performed MTT assays with different concentrations of gp120 and at 5Ā nM we did not observe cell toxicity. All experiments were conducted at 37Ā Ā°C in 5% CO2/95% air. Rosiglitazone, pioglitazone and GW9662 were dissolved in DMSO, and a total volume of 5Ā Ī¼L drug solution was added to 5Ā mL media in T-25 flasks. This volume of DMSO results in a final concentration of 0.001% DMSO. In order to keep conditions consistent between all treatments, we used the same concentration of DMSO in all flasks (e.g., control, vehicle gp120). Cells were treated with PPARĪ³ agonists (1Ā Ī¼M) rosiglitazone or (20Ā Ī¼M) pioglitazone in conjunction with 5Ā nM gp120. In order to demonstrate specificity of the PPARĪ³ agonists, cells were co-treated with PPARĪ³ specific antagonist GW9662 (500nM). For PPARĪ³ agonists, the doses were selected based on the EC50 values, rosiglitazone, 30ā€“100Ā nM, and pioglitazone, 500ā€“600Ā nM (Cayman Chemicals, Ann Arbor, Michigan, USA). Cell suspensions were collected 3 or 6Ā h after gp120 exposure and prepared for qPCR or immunoblotting analysis as described below.

Cell viability assay

Cell viability was assessed in primary cultures of rodent glial cells treated with HIV-1ADA gp120 using a standard MTT assay previously described by our laboratory [6]. In brief, cells were plated in 96-well assay plate at a density of 105/well. After 6Ā h, the medium was aspirated and replaced with fresh medium containing appropriate concentrations of DMSO, gp120, rosiglitazone, or pioglitazone (Additional file 1: Figure S1). These cultures were then incubated for 6Ā h; the medium was aspirated and replaced with fresh medium containing 10% of MTT (5.0Ā ng/mL). After 2h incubation, MTT solution was aspirated and 100Ā Ī¼L of DMSO was added to each well. The formazan content of each well was determined by UV spectrophotometry (570Ā nM) using a SpectraMax 384 microplate reader (Molecular Devices, Sunnyvale, CA).

Real-time quantitative polymerase chain reaction (qPCR)

Real-time quantitative polymerase chain reaction (qPCR) was applied to determine the transcript levels of inflammatory and oxidative stress markers, PPARĪ³ and GLT-1 according to previously published protocols by our laboratory [8]. Briefly, total RNA was extracted from cell culture or brain regions using TRizol reagent. The concentration of RNA was quantified spectrophotometrically by measuring absorbance at 260Ā nm. Extracted RNA (2000Ā ng) was treated with amplification grade DNase I to remove contaminating genomic DNA. The high capacity cDNA reverse transcriptase kit was used to synthesize first-strand cDNA. Rat primers were purchased from ThermoFisher Scientific (Waltham, MA, USA) for the following genes using TaqMan technology: Il-1Ī² (Rn00580432_m1), TNFĪ± (Rn99999017_m1), iNOS (Rn99999069_m1), PPARĪ³ (Rn00440945_m1), GLT-1 (Rn01486045_m1), and cyclophilin B (housekeeping gene; Rn 0835638_m1). Expression levels were normalized to housekeeping gene, cyclophilin B, and compared to saline-treated control group using the comparative Ct (Ī”Ī”Ct) method.

Immunoblot analysis

Western blot analysis was applied according to our previously published protocols to determine the protein expression of PPARĪ³, phosphorylated forms of NF-ĪŗB and GLT-1 [8]. In brief, cell culture and brain tissue homogenates were prepared using a modified RIPA lysis buffer (1% (v/v) NP-40 in 50Ā mM tris pHĀ 7.5, 150Ā mM NaCl, 1Ā mM ethylene glycol tetraacetic acid (EGTA), 1Ā mM sodium o-vanadate, 0.25% (v/v) sodium deoxycholic acid (Doc), 0.1% (v/v) sodium dodecyl sulfate (SDS), 200Ā Ī¼M phenylmethanesulfonyl fluoride, and 0.1% (v/v) protease inhibitor cocktail). Samples were sonicated for 10Ā s and centrifuged at 14,000Ā rpm for 15Ā min at 4Ā Ā°C to remove cellular debris. Nuclear extracts from rat hippocampus were prepared using a nuclear extraction kit from Abcam Inc. (Boston, MA, USA). The extracts were prepared as per the manufacturerā€™s protocol. Total protein (50Ā Ī¼g) was separated on 10% sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) and transferred onto a polyvinylidene difluoride membrane. After blocking with 5% skim milk for 2Ā h, the membrane was probed for protein of interest with primary antibody (rabbit polyclonal anti-PPARĪ³, 1:1000; rabbit polyclonal anti-phospho-p65 NFĪŗB, 1:100; or rabbit polyclonal anti-EAAT2 which recognizes residues 550 to C-terminus of rat glutamate transporter, 1:1000), and Ī²-actin was used as loading control (mouse monoclonal C4 anti-actin, 1:5000). HRP-conjugated secondary antibody was added after washes in tris-buffered saline with Tween. After further washing, bands were detected using enhanced chemiluminescent reagent. Densitometric analysis was performed in AlphaDigiDoc RT2 software (Alpha Innotech, San Leandro, CA, USA) to quantify relative protein expression. The graphs represent relative density of the bands of interest normalized to corresponding Ī²-actin and calculated fold changes based on control treated group.

Animals

Adult Wistar male rats, 250ā€“300Ā g, were purchased from Charles River Laboratories (St. Constant, Quebec, Canada) and were housed at the University of Toronto Division of Comparative Medicine with rodent chow and water on a 12-h light-dark cycle. All procedures were carried out in accordance with the approval of the University of Toronto Animal Care Committee. The rats were randomly assigned to four different groups: saline, gp120, rosiglitazone + gp120, and rosiglitazone + GW9662 + gp120, each group nĀ =Ā 6ā€“12 animals.

Animal surgery and intracerebralventricular (ICV) administration of HIV-1ADA gp120

Sterile stereotaxic technique was performed for all rat brain injections as previously described by our group [8]. In brief, 2ā€“5% isoflurane was used to induce surgical anesthesia. Prior to ICV, animals were administered subcutaneously ketoprofen (5Ā mg/kg) to induce analgesic effect. HIV-1ADA gp120-associated brain inflammation anesthetized rats were administered a single bilateral ICV injection of HIVADA gp120 (4Ā Ī¼g/4Ā Ī¼L/ventricle at a rate of 1Ā Ī¼L/min and sacrificed 6, 24, and 72Ā h post injection. A 5-Ī¼L Hamilton syringe was used to inject bilaterally into both ventricles at the following coordinates according to the Atlas of Paxinos and Watson (1986) 0.5Ā mm posterior to bregma, 1.5Ā mm lateral from midline, and 3.5Ā mm ventral from the surface of the skull. Control animals received an equal volume of saline. We have previously demonstrated the specificity of the gp120-mediated inflammatory response through the use of additional controls, (heat-inactivated gp120 and the CCR5 chemokine antagonist maraviroc) [8]. At the designated time points (6, 24, and 72Ā h) following ICV injection, animals were anesthetized and perfused through the left ventricle of the heart with a 240Ā mL phosphate buffered saline (PBS). At these time points, brain regions (hippocampus, frontal cortex, and striatum) were collected and harvested for further molecular and biochemical analysis. Samples were flash frozen in liquid nitrogen and kept at āˆ’80Ā Ā°C.

Intraperitoneal administration of rosiglitazone and GW9662

Animals (nĀ =Ā 6ā€“12 per group) were administered through intraperitoneal route (IP), 30Ā min prior to HIV-1ADA gp120 ICV with rosiglitazone (PPARĪ³ agonist; 10Ā mg/kg) or co-administration of rosiglitazone with GW9662 (PPARĪ³ antagonist; 5Ā mg/kg). Both compounds were dissolved in DMSO/saline 1:10. Saline (control) and gp120 (vehicle) animals received the same volume of DMSO/saline 1:10 IP. These compounds are known to effectively permeate across the BBB [31]. The dose selected for rosiglitazone (10Ā mg/kg) was chosen based on previous reports which demonstrated the neuroprotective effects of rosiglitazone in vivo [23, 25, 26, 31].

Data analysis

Studentā€™s t test was used to determine statistical significance between two groups. Multiple comparisons were performed using one-way ANOVA with Bonferroniā€™s post-hoc analysis. A p value less than 0.05 was considered statistically significant. Data was analyzed using GraphPad Prism software (San Diego, CA). Each set of in vitro experiments were repeated at least three times in cells pertaining to different isolations, and for the in vivo experiments, samples were collected from 4 to 12 animals per group.

Results

PPARĪ³ agonists rosiglitazone and pioglitazone reverse HIV-1ADA gp120-mediated inflammatory responses in vitro, in primary cultures of rat mixed astrocytes and microglia

Previous studies from our laboratory have shown that exposure to HIV-196ZM651 gp120 induces mRNA expression of pro-inflammatory cytokines and oxidative stress markers in primary cultures of astrocytes [6]. Herein, we confirm this inflammatory response using an additional strain of gp120 (ADA) in primary cultures of mixed astrocytes and microglia. These cultures have been characterized through immunocytochemical staining for astrocyte specific marker GFAP and microglia specific marker cd11b/c (Fig. 1). Exposure of the cells to HIV-1ADA gp120 (5Ā nM) significantly increased the inflammatory markers (TNFĪ± and IL-1Ī²) and indicator of oxidative stress responses (iNOS) at 3Ā h post gp120 exposure. Treatment with rosiglitazone (1Ā Ī¼M) or pioglitazone (20Ā Ī¼M) significantly reversed the inflammatory responses (Fig.Ā 2). Other doses for rosiglitazone (250 and 500Ā nM) were tested; however, the 1-Ī¼M dose appeared to be the most effective (Additional file 2: Figure S2). For pioglitazone, doses of 1 and 50Ā Ī¼M were also examined, and the 1Ī¼M dose appeared too low to exhibit a significant anti-inflammatory effect (Additional file 2: Figure S2). To confirm that the anti-inflammatory effects of PPARĪ³ agonists rosiglitazone and pioglitazone were PPARĪ³ dependent, cells were co-administered with the PPARĪ³ specific antagonist GW9662. As expected, we observed that GW9662 (500Ā nM) abolished the effects of both agonists (Fig. 2). We performed an MTT assay in primary cultures of mixed astrocytes and microglia to verify that the treatments did not significantly alter cell proliferation and viability; in all cases, cell viability was not significantly different from control (i.e., untreated) cultures (Additional file 1: Figure S1).

Fig. 2
figure 2

PPARĪ³ agonists rosiglitazone and pioglitazone reverse HIV-1ADA gp120-mediated inflammatory responses in vitro. Primary cultures of mixed rat astrocytes and microglia were treated with PPARĪ³ agonists, rosiglitazone (1Ā Ī¼M) or pioglitazone (20Ā Ī¼M) or co-treated ļ»æwith ļ»æPPARĪ³ antagonist GW9662 (500nM)ļ»æ for 1Ā h prior to gp120 (5Ā nM) exposure for 3Ā h, and a TNF-Ī±, b IL-1Ī², and c iNOS mRNA levels were measured using qPCR. Cyclophilin BĀ was used as the housekeeping gene. Results are expressed as meanĀ Ā±Ā SEM relative to the DMSO (control) of at least 3 separate experiments. Asterisks and pound symbol represent data points significantly different from DMSO (control) and gp120 (vehicle) respectively (*pĀ <Ā 0.05, **pĀ <Ā 0.01, ***pĀ <Ā 0.001, ****pĀ <Ā 0.0001, #pĀ <Ā 0.05, ##pĀ <Ā 0.01) (aā€“c)

PPARĪ³ agonist rosiglitazone reverses gp120-mediated inflammatory responses in vivo, in an HIV-1ADA gp120 ICV-administered rodent model

In our present study, the dose of gp120 for ICV administration (4Ā Ī¼g/ventricle) was chosen based on previous reports using a similar range of doses (1ā€“4Ā Ī¼g/ventricle) injected into the rodent brain to induce an inflammatory effect [32]. In our hands, a single dose of HIV-1ADA gp120 (4Ā Ī¼g/ventricle) induced a significant increase in inflammatory and indicator of oxidative stress responses (TNFĪ±, IL-1Ī², and iNOS) at 24Ā h in the hippocampus (Fig.Ā 3) and frontal cortex (Additional file 3: Figure S3).

Fig. 3
figure 3

PPARĪ³ agonist rosiglitazone reverses HIV-1ADA gp120-mediated inflammatory responses in hippocampus. Adult Wistar rats were administered IP, 30Ā min prior to ICV bilateral injection of 4Ā Ī¼g/ventricle HIV-1ADA gp120 with rosiglitazone (10Ā mg/kg) or co-administration of rosiglitazone with GW9662 (5Ā mg/kg). Saline (control) and gp120 (vehicle) animals received the same volume of DMSO/saline 1:10 IP. Hippocampus brain regions were isolated 24Ā h post ICV, and a TNFĪ±, b IL-1Ī², and c iNOS mRNA levels were measured using qPCR. Cyclophilin BĀ was used as the housekeeping gene. Results are expressed as meanĀ Ā±Ā SEM relative to saline group (control) nĀ =Ā 7ā€“12 animals/group. Asterisks and pound symbol represent data points significantly different from saline (control) and gp120 (vehicle) respectively (*pĀ <Ā 0.05, **pĀ <Ā 0.01, ****pĀ <Ā 0.0001, ##pĀ <Ā 0.01)

To evaluate whether PPARĪ³ agonists protect against gp120-induced expression of inflammatory genes TNFĪ±, IL-1Ī², and indicator of oxidative stress response iNOS, animals administered with gp120 (4Ā Ī¼g/ventricle) were treated with or without an IP dose of 10Ā mg/kg rosiglitazone. Treatment with PPARĪ³ specific agonist rosiglitazone attenuated gp120-induced expression of TNFĪ± and IL-1Ī². Although a trend towards reduced expression of iNOS was evident with rosiglitazone treatment, this result did not reach statistical significance (Fig. 3). In order to investigate the specificity of PPARĪ³ mediating the protective effects of rosiglitazone, animals were co-administered with an IP dose of 5Ā mg/kg PPARĪ³ specific antagonist GW9662. The administration of the antagonist abolished the effects of rosiglitazone in reducing levels of TNFĪ±, IL-1Ī², and iNOS (Fig. 3). Similar effects were observed in the frontal cortex brain region (Additional file 3: Figure S3).

PPARĪ³ agonists reverse gp120-mediated downregulation of GLT-1 in vitro, in primary cultures of rat astrocytes, and in vivo, in an HIV-1ADA gp120 ICV-administered rodent model

Several studies have demonstrated that the glutamate transporter EAAT2 (human) or GLT-1 (rodent) is downregulated in the context of neurological disorders [18, 33]. In particular, gp120 exposure has been reported to decrease functional expression of excitatory amino acid transporter 2 (EAAT2) in primary cultures of human astrocytes [17]. Therefore, in our in vitro and in vivo systems of HIV-1ADA gp120-associated brain inflammation, we investigated levels of the rodent homolog, GLT-1 (Fig.Ā 4). In primary cultures of rat astrocytes, gp120 exposure for 6Ā h resulted in a significant downregulation of GLT-1 at the protein level, and treatment with PPARĪ³ agonists rosiglitazone (1Ā Ī¼M) or pioglitazone (20Ā Ī¼M) significantly restores the levels (Fig. 4a). This was also observed, in vivo, in the HIV-1ADA gp120 ICV-administered rodent model where we found a significant downregulation of GLT-1 at the protein level 24Ā h post ICV (Fig. 4b). We were able to further demonstrate that treatment with rosiglitazone restored mRNA levels of GLT-1 in the hippocampus 24Ā h post ICV (Fig. 4c). Animals were co-administered PPARĪ³ antagonist GW9662 (5Ā mg/kg) in order to investigate the specificity of PPARĪ³ mediating the protective effects of rosiglitazone. GW9662 administration abolished the effects of rosiglitazone, demonstrating that the restored levels of GLT-1 with rosiglitazone treatment were likely mediated through PPARĪ³ (Fig. 4c).

Fig. 4
figure 4

PPARĪ³ agonists reverse HIV-1ADA gp120-mediated downregulation of GLT-1 in vitro and in vivo. Primary cultures of rat astrocytes were treated with PPARĪ³ ligands 1Ā h prior to gp120 (5Ā nM) exposure for 6Ā h, and protein expression of GLT-1 was analyzed through immunoblotting (a). Adult Wistar rats were administered IP, 30Ā min prior to ICV bilateral injection of 4Ā Ī¼g/ventricle HIV-1ADA gp120 with rosiglitazone (10Ā mg/kg) or co-administration of rosiglitazone with GW9662 (5Ā mg/kg). Saline (control) and gp120 (vehicle) animals received the same volume of DMSO/saline 1:10 IP. Hippocampus brain regions were isolated 24Ā h post ICV; protein expression of GLT-1 was analyzed through immunoblotting (b), and GLT-1 mRNA levels were measured using qPCR (c). For immunoblotting, cell or tissue protein lysate (50Ā Ī¼g) was resolved on a 10% SDS-polyacrylamide gel, transferred to a PVDF membrane. GLT-1 was detected using a rabbit polyclonal antibody (1:1000, dilution). Actin was detected using a mouse monoclonal antibody (1:5000, dilution). Data generated from densitometric analysis is presented as a ratio of GLT-1 expression normalized to actin (loading control). Cyclophilin B was used as the housekeeping gene for qPCR. Results are expressed as meanĀ Ā±Ā SEM relative to DMSO (control, in vitro) or saline (control, in vivo) of at least 3 separate experiments in vitro and nĀ =Ā 5ā€“7 animals/group in vivo. Asterisks and pound symbol represent data points significantly different from DMSO or saline (control) and gp120 (vehicle) respectively (**pĀ <Ā 0.01, *pĀ <Ā 0.05, #pĀ <Ā 0.05)

PPARĪ³ is downregulated in response to gp120 in vitro, in primary cultures of rat astrocytes, and in vivo, in an HIV-1ADA gp120 ICV-administered rodent model

It has been reported that expression of PPARĪ³ can be altered in response to HIV-1 or other inflammatory stimuli [34, 35]. Therefore, we also investigated the expression of PPARĪ³. In vitro, in primary cultures of rat astrocytes, we observed a significant downregulation of PPARĪ³ at the mRNA level 6Ā h post gp120 exposure (Fig.Ā 5a) with a similar trend evident in the primary cultures of mixed glial cells exposed to gp120 for 3Ā h (Fig. 5b). In vivo, mRNA levels of PPARĪ³ were examined at 6Ā h post ICV, and a significant decrease was also observed in the hippocampus (Fig. 5c). At a later time point, we also investigated the corresponding protein levels and observed a profound downregulation 72Ā h post ICV (Fig. 5d). Similar data were obtained in the frontal cortex (Additional file 4: Figure S4).

Fig. 5
figure 5

Effect of HIV-1ADA gp120 on the protein and mRNA expression of PPARĪ³ in vitro and in vivo. Primary cultures of rat astrocytes were exposed to g120 (5Ā nM) for 6Ā h, and PPARĪ³ mRNA levels were measured using qPCR (a). Primary cultures of mixed rat astrocytes and microglia were exposed to g120 (5Ā nM) for 3Ā h, and PPARĪ³ mRNA levels were measured using qPCR (b). Adult Wistar rats were administered, bilateral ICV, 4Ā Ī¼g/ventricle of gp120; hippocampus was isolated 6ā€“72Ā h post ICV, and PPARĪ³ mRNA levels were measured using qPCR (c), and protein expression of PPARĪ³ was analyzed through immunoblotting (d). Cyclophillin was used as the housekeeping gene for qPCR. For immunoblotting, hippocampus tissue protein lysates (50Ā Ī¼g) were resolved on a 10% SDS-polyacrylamide gel and transferred to a PVDF membrane. HepG2 (50Ā Ī¼g) were used as positive control for PPARĪ³ protein. PPARĪ³ was detected using a rabbit polyclonal PPARĪ³ antibody (1:1000 dilution). pt?>Actin was detected using a mouse monoclonal antibody (1:5000, dilution). Data generated from densitometric analysis is presented as a ratio of PPARĪ³ expression normalized to actin (loading control). Results are expressed as meanĀ Ā±Ā SEM relative to DMSO (control, in vitro) or saline (control, in vivo) of at least 3 separate experiments in vitro and nĀ =Ā 6ā€“11 animals/group in vivo. Asterisks represent data point significantly different from DMSO or saline (control) (****pĀ <Ā 0.0001, *pĀ <Ā 0.05)

Involvement of NF-ĪŗB redox regulated transcription factor

In order to investigate the involvement of the transcriptional factor NF-ĪŗB, animals were administered HIV-1ADA gp120 ICV and rosiglitazone IP but sacrificed at an earlier time point post ICV injection (5Ā h). The hippocampus tissue was extracted for nuclear proteins, and the phosphorylated levels of the p-65 subunit of NF-ĪŗB were analyzed by Western blot. The phosphorylated forms of p-65 correspond to its activation in the nucleus. Our data showed that treatment with rosiglitazone decreased the gp120-induced activation of p-65 in the hippocampus (Fig.Ā 6).

Fig. 6
figure 6

PPARĪ³ agonists reverse HIV-1ADA gp120-induced NF-ĪŗB (p-p65) phosphorylation in vivo. Adult Wistar rats were administered IP, 30Ā min prior to ICV bilateral injection of 4Ā Ī¼g/ventricle HIV-1ADA gp120 with rosiglitazone (10Ā mg/kg). Saline (control) and gp120 (vehicle) animals received the same volume of DMSO/saline 1:10 IP. Hippocampus brain regions were isolated 5Ā h post ICV. For immunoblotting, hippocampus tissue nuclear extracts (50Ā Ī¼g) were resolved on a 10% SDS-polyacrylamide gel and transferred to a PVDF membrane. K-562 (immortalized myelogenous leukemia cell line) was used as a positive control for p-p65 protein. NF-ĪŗB (p-p65) was detected using a rabbit polyclonal p-p65Ser536 (1:100 dilution). Actin was detected using a mouse monoclonal antibody (1:5000, dilution). Data generated from densitometric analysis is presented as a ratio of p-p65Ser536 expression normalized to actin (loading control). Results are expressed as meanĀ Ā±Ā SEM relative to saline group (control) nĀ =Ā 3ā€“4 animals/group. Asterisks and pound symbol represent data points significantly different from saline (control) and gp120 (vehicle) respectively (***pĀ <Ā 0.001, ###pĀ <Ā 0.001)

Discussion

The limitations of currently used ARVs include poor brain penetration, lack of direct anti-inflammatory properties, and neurotoxicity associated with better permeable drugs. Identifying therapeutic compounds that can effectively permeate the BBB and exhibit anti-inflammatory properties may provide an option in treating or preventing HAND.

In this study, we implemented an in vitro model of gp120-associated inflammation using primary cultures of mixed rat astrocytes and microglia exposed to 5Ā nM R5-tropic HIV-1ADA gp120. R5-tropic strains are considered to be the most prevalent in the brain as the CCR5 co-receptor is expressed on a broad spectrum of cells in the CNS such as microglia, astrocytes, and neurons [36]. The concentration of gp120 in the brain of HIV-1 patients is not clearly documented for obvious reasons of tissue limitation; however, in the periphery (i.e., serum), concentrations of gp120 have been reported to be as high as 92Ā ng/mL [37]. Immunohistochemical analysis has also detected gp120 in brain tissue from HIV-1-infected patients [38]. Furthermore, studies by Banks and Kastin showed that gp120 crosses the mouse blood brain barrier (BBB) after I.V. administration and the transport is likely mediated through lectin-like mechanisms resembling adsorptive endocytosis. Two hours post I.V. injection of gp120, the percent uptake was 0.15% per gram of brain [39]. Our group has previously demonstrated gp120-mediated inflammatory response in vitro, in primary cultures of rodent and human astrocytes, as well as, in vivo in a rodent model of ICV administered HIV-1ADA gp120 [6,7,8, 40]. Herein, we sought to identify molecular pathways that may play a role in decreasing the gp120-induced acute inflammatory response. PPARĪ³ agonists have been widely used for the treatment of type II diabetes; however, they are also known to exhibit anti-inflammatory and anti-oxidant properties in several models of CNS disorders [24]. It is important to note that although previous reports from randomized clinical trials of rosiglitazone suggested an elevated risk of cardiovascular toxicity, data from the 2009 RECORD trial, a six-year open label randomized control trial with 4447 patients, failed to show risk of overall cardiovascular mortality and morbidity in comparison with other standard type II diabetes medications (metformin, sulfonyl-urea) [41]. In light of these findings, the FDA has removed restrictions from rosiglitazone. Furthermore, we are also using pioglitazone, another PPARĪ³ ligand, which has shown to have reduced cardiovascular risks [42]. To date, a few studies have reported anti-inflammatory potential of PPARĪ³ agonists in attenuating HIV-associated inflammatory response [5, 26, 43]. Although it has been reported that rosiglitazone treatment in primary cultures of rodent astrocytes attenuated LPS-induced secretion of several pro-inflammatory markers (IL-12, TNFĪ±, IL-1Ī², IL-6, MCP-1 [44, 45], this effect has not been thoroughly examined in astrocytes or microglia in the context of gp120-associated pathologies. Our in vitro data demonstrated that glial treatment with either rosiglitazone or pioglitazone reversed the gp120-mediated inflammatory responses. Furthermore, we showed that co-administration of a synthetic PPARĪ³ antagonist, GW9662, which acts as a potent, irreversible, and selective PPARĪ³ antagonist by modifying a cysteine residue in the ligand-binding site of PPARĪ³, abolished the rosiglitazone or pioglitazone anti-inflammatory effects, suggesting that these effects are specifically mediated by PPARĪ³.

Next, we sought to characterize an acute in vivo model of HIV-1-associated brain inflammation by ICV administration of HIV-1ADA gp120. A single bilateral ICV dose of (4Ā Ī¼g/ventricle) resulted in a significant induction of inflammatory genes (TNFĪ±, IL-1Ī², iNOS) at 24Ā h in the hippocampus (Fig. 3) and frontal cortex (Additional file 3: Figure S3). Our group has previously demonstrated that this inflammatory response is mediated through direct interaction of R5-tropic gp120 with chemokine co-receptor CCR5 in vitro and in vivo [6, 8].

We then investigated the anti-inflammatory properties of rosiglitazone in our in vivo rodent model of HIV-1ADA gp120-induced brain inflammation. Our results showed that treatment with 10Ā mg/kg rosiglitazone reduced gp120-induced gene expression of inflammatory markers (TNFĪ± and IL-1Ī²) in hippocampus (Fig. 3). These results are in agreement with other studies that have observed rosiglitazone-mediated downregulation of inflammatory cytokines in various in vivo models of CNS disorders [23]. To the best of our knowledge, only two studies have examined the in vivo therapeutic efficacy of PPARĪ³ agonists in the context of HIV-1 infection in the brain. Huang et al. showed that rosiglitazone treatment reduced HIV-1 viral protein Tat increase in BBB permeability, astrogliosis, and neuronal loss [5]. Potula et al. used a severe combined immunodeficiency mouse model of HIV-1 encephalitis, where treatment of rosiglitazone resulted in suppression of viral replication in brain macrophages [25]. The anti-inflammatory effects of rosiglitazone in our gp120 model were abolished with co-administration of 5Ā mg/kg GW9662 (PPARĪ³ antagonist), demonstrating that the anti-inflammatory effects are specifically mediated by PPARĪ³ (Fig. 3).

Glutamate is the most abundant neurotransmitter in mammalian CNS. Clearance of glutamate from the extracellular space is regulated by specific uptake transporters existing in the plasma membrane of glial cells and neurons. Many cell types throughout the brain express glutamate transporters; however, uptake by astrocytes is quantitatively the most significant [46]. EAAT2/GLT-1 is ļ»æthe primary transporter ļ»æ responsible for glutamate uptake in the mammalian brain, and impairment to this transporter can result in glutamate excitotoxicity which has been proposed to contribute to several neurological diseases including HAND [47]. It has been reported that HIV-1-infected individuals have five-fold greater levels of glutamate in the CSF compared to healthy controls [48]. More recently, studies investigating glutamate levels in patients receiving combinational antiretroviral therapy observed selective increases in glutamate CSF levels in patients with HAND compared to those without neurological impairments [16]. It has been proposed that gp120 may play an important role in regulating the glutamate transporter by decreasing its functional expression in primary cultures of human astrocytes [17] and in gp120 transgenic mice [18]. In addition, others have reported that gp120 may be disrupting ion fluxes across the plasma membrane [49] or stimulating glutamate release [19]. Our data corroborate those of other groups who have demonstrated decreased expression of GLT-1 after gp120 exposure [17, 18]. This effect has been proposed to be mediated by TNFĪ± which could transcriptionally repress GLT-1 through activation of the NF-ĪŗB pathway [50].

Strategies to modulate glutamate excitotoxicity include the use of memantine, an N-methyl-d-aspartate (NMDA) receptor antagonist; however, initial clinical trials in patients with HAND were unsuccessful [51]. Other on-going strategies include regulation of enzymes that are responsible for producing glutamate [52] or transporters involved with glutamate release [53]; however, currently, no clinically available brain-penetrating compounds exist. To date, a limited number of studies have investigated PPARĪ³ as a target for GLT-1 regulation [33, 54]. In light of these recent reports demonstrating that PPARĪ³ activation increases astrocytic GLT-1 expression in the context of ischemia and glioma cells, we showed that targeting PPARĪ³ for GLT-1 modulation is also applicable in the context of HIV-1-associated brain inflammation. The mechanism for restored GLT-1 expression could be due to the anti-inflammatory effects of these agonists which we have shown to attenuate TNFĪ± release and activation of the NF-ĪŗB pathway (Fig. 6). In addition, previous bioinformatic analyses revealed that there are at least 6 putative consensus PPAR response element sites in the promoter region of the EAAT2 gene and in vitro treatment with rosiglitazone increased promoter activity [33].

We also examined PPARĪ³ expression following gp120 administration. A profound downegulation of PPARĪ³ was observed at both the mRNA and protein levels after gp120 exposure (Fig. 5). Similar downregulatory effects on PPARĪ³ has previously been shown in other tissues and cell systems of inflammatory diseases [34, 55]. Furthermore, downregulation of this nuclear receptor has been demonstrated in lung tissue in the context of HIV-1-associated interstitial pneumonitis [56] and chronic obstructive pulmonary disease [57]. Recently, immunohistochemical analysis of feline immunodeficiency virus animal brains and HIV-1-infected post-mortem brain tissue revealed a reduced expression of PPARĪ³ [35]. In addition, it has also been reported that PPARĪ³ can be downregulated through activation of the mitogen-activated kinases such as extracellular signal-regulated kinases 1 and 2 (ERK1/2) which can phosphorylate the activation function resulting in negative feedback [58]. We have previously demonstrated that this pathway is activated in response to gp120 [8]. Together, these studies including our own suggest the involvement of this nuclear receptor in HIV-1-mediated inflammatory response.

To investigate the mechanisms which could be involved in the PPAR-mediated anti-inflammatory effects, we examined the effect of rosiglitazone treatment on the suppression of redox regulated transcriptional factor NF-ĪŗB upregulated by gp120. NF-ĪŗB binding sites have been reported in several promoter regions of inflammatory cytokine genes [59], and two binding sites have also been identified in the promoter-proximal enhancer region of HIV-1 LTR [60]. Several mechanisms have been reported for the PPARĪ³-mediated inhibition of NF-ĪŗB; these mechanisms include physical interaction of PPARĪ³ with NF-ĪŗB, co-activator competition of both transcriptional factors regulation of protein localization, and prevention of signal dependent clearance of co-repressor complexes on inflammatory promoters [28, 29]. Our data suggest that treatment with PPARĪ³ agonist rosiglitazone in vivo decreases significantly the gp120-induced phosphorylation of NF-ĪŗB in the hippocampus.

Conclusion

Findings from our in vitro and in vivo work revealed that PPARĪ³ is an important pathway involved in HIV-1 brain-associated inflammation and could constitute a potential molecular target in the treatment/prevention of HIV-1 brain inflammation and HAND.

Abbreviations

ARVs:

Antiretroviral drugs

BBB:

Blood-brain barrier

CNS:

Central nervous system

CSF:

Cerebrospinal fluid

DAPI:

4ā€²,6ā€²-diamidino-2-phenylindole hydrochloride

Doc:

Deoxycholic acid

EAAT2:

Excitatory amino acid transporter 2

EGTA:

Ethylene glycol tetraacetic acid

ERK:

Extracellular regulated kinase

GLT-1:

Glutamate transporter 1

gp120:

Glycoprotein 120

GW9662:

2-Chloro-5-nitro-N-phenylbenzamide

HAART:

Highly active antiretroviral therapy

HAND:

HIV-associated neurocognitive disorder

HIV:

Human immunodeficiency virus

ICV:

Intracerebralventricular

IL-1Ī²:

Interleukin-1beta

IL-6:

Interleukin-6

IL-8:

Interleukin-8

iNOS:

Inducible nitric oxide synthase

IP:

Intraperitoneal

LPS:

Lipopolysaccharide

NF-ĪŗB:

Nuclear factor kappa B

NO:

Nitric oxide

PBS āˆ’/āˆ’:

Phosphate-buffered saline without Ca2+ and Mg+

PPARĪ³:

Peroxisome proliferator-activated receptor gamma

ROS:

Reactive oxygen species

SDS:

Sodium dodecyl sulfate

Tat:

Transactivator of transcription

TNFĪ±:

Tumor necrosis factor alpha

Vpr:

Viral protein R

References

  1. Liu NQ, Lossinsky AS, Popik W, Li X, Gujuluva C, Kriederman B, et al. Human immunodeficiency virus type 1 enters brain microvascular endothelia by macropinocytosis dependent on lipid rafts and the mitogen-activated protein kinase signaling pathway. J Virol. 2002;76:6689ā€“700.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  2. Dohgu S, Ryerse JS, Robinson SM, Banks WA. Human immunodeficiency virus-1 uses the mannose-6-phosphate receptor to cross the blood-brain barrier. PLoS One. 2012;7:1ā€“12.

    ArticleĀ  Google ScholarĀ 

  3. Lindl KA, Marks DR, Kolson DL, Jordan-Sciutto KL. HIV-associated neurocognitive disorder: pathogenesis and therapeutic opportunities. J NeuroImmune Pharmacol. 2010;5:294ā€“309.

    ArticleĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  4. Kaul M, Garden GA, Lipton SA. Pathways to neuronal injury and apoptosis in HIV-associated dementia. Nature. 2001;410:988ā€“94.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  5. Huang W, Chen L, Zhang B, Park M, Toborek M. PPAR agonist-mediated protection against HIV Tat-induced cerebrovascular toxicity is enhanced in MMP-9-deficient mice. J Cereb Blood Flow Metab. 2014;34:646ā€“53.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  6. Ronaldson PT, Bendayan R. HIV-1 viral envelope glycoprotein gp120 triggers an inflammatory response in cultured rat astrocytes and regulates the functional expression of P-glycoprotein. Mol Pharmacol. 2006;70:1087ā€“98.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  7. Ronaldson PT, Bendayan R. HIV-1 viral envelope glycoprotein gp120 produces oxidative stress and regulates the functional expression of multidrug resistance protein-1 (Mrp1) in glial cells. J Neurochem. 2008;106:1298ā€“313.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  8. Ashraf T, Jiang W, Hoque MT, Henderson J, Wu C, Bendayan R. Role of anti-inflammatory compounds in human immunodeficiency virus-1 glycoprotein120-mediated brain inflammation. J Neuroinflammation. 2014;11:91.

    ArticleĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  9. Saylor D, Dickens AM, Sacktor N, Haughey N, Slusher B, Pletnikov M, et al. HIV-associated neurocognitive disorderā€”pathogenesis and prospects for treatment. Nat Rev Neurol. 2016;12:234ā€“48.

    ArticleĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  10. Vivithanaporn P, Heo G, Gamble J, Krentz HB, Hoke A, Gill MJ, et al. Neurologic disease burden in treated HIV/AIDS predicts survival: a population-based study. Neurology. 2010;75:1150ā€“8.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  11. Cavalcante GIT, Capistrano VLM, Cavalcante FSD, Vasconcelos SMM, MacĆŖdo DS, Sousa FCF, et al. Implications of efavirenz for neuropsychiatry: a review. Int J Neurosci. 2010;120:739ā€“45.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  12. Best BM, Letendre SL, Brigid E, Clifford DB, Collier AC, Gelman BB, et al. Low atazanavir concentrations in cerebrospinal fluid. AIDS. 2010;23:83ā€“7.

    ArticleĀ  Google ScholarĀ 

  13. Best BM, Letendre SL, Koopmans P, Rossi SS, Clifford DB, Collier AC, et al. Low CSF concentrations of the nucleotide reverse transcriptase inhibitor, tenofovir. AIDS. 2013;59:376ā€“81.

    Google ScholarĀ 

  14. Piacenti FJ. An update and review of antiretroviral therapy. Pharmacotherapy. 2006;26:1111ā€“33.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  15. Kamat A, Lyons J, Misra V. Monocyte activation markers in cerebrospinal fluid associated with impaired neurocognitive testing in advanced HIV infection. J Acquir Immune Defic Syndr. 2012;60:234ā€“43.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  16. Cassol E, Misra V, Dutta A, Morgello S, Gabuzda D. Cerebrospinal fluid metabolomics reveals altered waste clearance and accelerated aging in HIV patients with neurocognitive impairment. AIDS. 2014;28:1579ā€“91.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  17. Wang Z, Pekarskaya O, Bencheikh M, Chao W, Gelbard HA, Ghorpade A, et al. Reduced expression of glutamate transporter EAAT2 and impaired glutamate transport in human primary astrocytes exposed to HIV-1 or gp120. J Virol. 2003;312:60ā€“73.

    ArticleĀ  CASĀ  Google ScholarĀ 

  18. Melendez RI, Roman C, Capo-Velez CM, Lasalde-Dominicci JA. Decreased glial and synaptic glutamate uptake in the striatum of HIV-1 gp120 transgenic mice. J Neuro-Oncol. 2015;22:358ā€“65.

    Google ScholarĀ 

  19. Vesce S, Bezzi P, Rossi D, Meldolesi J, Volterra A. HIV-1 gp120 glycoprotein affects the astrocyte control of extracellular glutamate by both inhibiting the uptake and stimulating the release of the amino acid. FEBS Lett. 1997;411:107ā€“9.

  20. Grygiel-GĆ³rniak B. Peroxisome proliferator-activated receptors and their ligands: nutritional and clinical implicationsā€”a review. Nutr J. 2014;13:17.

    ArticleĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  21. Polak PE, Kalinin S, Dello Russo C, Gavrilyuk V, Sharp A, Peters JM, et al. Protective effects of a peroxisome proliferator-activated receptor-beta/delta agonist in experimental autoimmune encephalomyelitis. J Neuroimmunol. 2005;168:65ā€“75.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  22. Barbiero JK, Santiago R, Tonin FS, Boschen S, da Silva LM, Werner MF, et al. PPAR-Ī± agonist fenofibrate protects against the damaging effects of MPTP in a rat model of Parkinsonā€™s disease. Prog Neuro-Psychopharmacol Biol Psychiatry. 2014;53:35ā€“44.

    ArticleĀ  CASĀ  Google ScholarĀ 

  23. Shao Z-Q, Liu Z-J. Neuroinflammation and neuronal autophagic death were suppressed via Rosiglitazone treatment: new evidence on neuroprotection in a rat model of global cerebral ischemia. J Neurol Sci. 2015;349:65ā€“71.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  24. Bernardo A, Minghetti L. Regulation of glial cell functions by PPAR-gamma natural and synthetic agonists. PPAR Res. 2008;2008.

  25. Potula R, Ramirez SH, Knipe B, Leibhart J, Heilman D, Morsey B, et al. Peroxisome proliferator-activated receptor-gamma activation suppresses HIV-1 replication in an animal model of encephalitis. AIDS. 2009;22:1539ā€“49.

  26. Huang W, Eum SY, AndrĆ”s IE, Hennig B, Toborek M. PPARalpha and PPARgamma attenuate HIV-induced dysregulation of tight junction proteins by modulations of matrix metalloproteinase and proteasome activities. FASEB J. 2009;23:1596ā€“606.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  27. Bernier A, Cleret-buhot A, Zhang Y, Goulet J, Monteiro P. Transcriptional profiling reveals molecular signatures associated with HIV permissiveness in Th1Th17 cells and identifies peroxisome proliferator-activated receptor gamma as an intrinsic negative regulator of viral replication. Retrovirology. 2013; 10:160.

  28. Sauer S. Ligands for the nuclear peroxisome proliferator-activated receptor gamma. Trends Pharmacol Sci. 2015;36:688ā€“704.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  29. Ricote M, Glass CK. PPARs and molecular mechanisms of transrepression. Biochim Biophys Acta - Mol Cell Biol Lipids. 2007, 1771:926ā€“35.

  30. Hoque MT, Robillard KR, Bendayan R. Regulation of breast cancer resistant protein by peroxisome proliferator-activated receptor Ī± in human brain microvessel endothelial cells. Mol Pharmacol. 2012;81:598ā€“609.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  31. Strum JC, Shehee R, Virley D, Richardson J, Mattie M, Selley P, et al. Rosiglitazone induces mitochondrial biogenesis in mouse brain. J Alzheimers Dis. 2007;11:45ā€“51.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  32. Pugh CR, Johnson JD, Martin D, Rudy JW, Maier SF, Watkins LR. Human immunodeficiency virus-1 coat protein gp120 impairs contextual fear conditioning: a potential role in AIDS related learning and memory impairments. Brain Res. 2000;861:8ā€“15.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  33. Romera C, Hurtado O, Mallolas J, Pereira MP, Morales JR, Romera A, et al. Ischemic preconditioning reveals that GLT1/EAAT2 glutamate transporter is a novel PPARgamma target gene involved in neuroprotection. J Cereb Blood Flow Metab. 2007;27:1327ā€“38.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  34. Tanaka T, Itoh H, Doi K, Fukunaga Y, Hosoda K, Shintani M, et al. Down regulation of peroxisome proliferator-activated receptor-gamma expression by inflammatory cytokines and its reversal by thiazolidinediones. Diabetologia. 1999;42:702ā€“10.

  35. Mamik MK, Asahchop EL, Chan WF, Zhu Y, Branton WG, McKenzie BA, et al. Insulin treatment prevents neuroinflammation and neuronal injury with restored neurobehavioral function in models of HIV/AIDS neurodegeneration. J Neurosci. 2016;36:10683ā€“95.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  36. Gabuzda D, Wang J. Chemokine receptors and mechanisms of cell death in HIV neuropathogenesis. J Neuro-Oncol. 2000;6:S24ā€“32.

    CASĀ  Google ScholarĀ 

  37. Oh SK, Cruikshank WW, Raina J, Blanchard GC, Adler WH, Walker J, et al. Identification of HIV-1 envelope glycoprotein in the serum of AIDS and ARC patients. J Acquir Immune Defic Syndr. 1992;5:251ā€“6.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  38. Jones MV, Bell JE, Nath A. Immunolocalization of HIV envelope gp120 in HIV encephalitis with dementia. AIDS. 2000;14:2709ā€“13.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  39. Banks WA, Kastin AJ. Characterization of lectin-mediated brain uptake of HIV-1 GP120. J Neurosci Res. 1998;54:522ā€“9.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  40. Ashraf T, Ronaldson PT, Persidsky Y, Bendayan R. Regulation of P-glycoprotein by human immunodeficiency virus-1 in primary cultures of human fetal astrocytes. J Neurosci Res. 2011;89:1773ā€“82.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  41. Home PD, Pocock SJ, Beck-Nielsen H, Curtis PS, Gomis R, Hanefeld M, et al. Rosiglitazone evaluated for cardiovascular outcomes in oral agent combination therapy for type 2 diabetes (RECORD): a multicentre, randomised, open-label trial. Lancet. 2009;373:2125ā€“35.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  42. Dormandy JA, Charbonnel B, Eckland DJ, Erdmann E, Massi-Benedetti M, Moules IK, et al. Secondary prevention of macrovascular events in patients with type 2 diabetes in the PROactive Study (PROspective pioglitazone clinical trial in macrovascular events): a randomised controlled trial. Lancet. 2005;366:1279ā€“89.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  43. Huang W, Rha GB, Han M-J, Eum SY, AndrĆ”s IE, Zhong Y, et al. PPARalpha and PPARgamma effectively protect against HIV-induced inflammatory responses in brain endothelial cells. J Neurochem. 2008;107:497ā€“509.

    ArticleĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  44. Storer PD, Xu J, Chavis J, Drew PD. Peroxisome proliferator-activated receptor-gamma agonists inhibit the activation of microglia and astrocytes: implications for multiple sclerosis. J Neuroimmunol. 2005;161:113ā€“22.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  45. Xu J, Drew PD. Peroxisome proliferator-activated receptor-gamma agonists suppress the production of IL-12 family cytokines by activated glia. J Immunol. 2007;178:1904ā€“13.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  46. Anderson CM, Swanson RA. Astrocyte glutamate transport: review of properties, regulation, and physiological functions. Glia. 2000;32:1ā€“14.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  47. Vazquez-Santiago F, Noel R Jr, Porter JT, Rivera-Amill V. Glutamate metabolism and HIV-associated neurocognitive disorders. J Neuro-Oncol. 2014;20:315ā€“31.

    CASĀ  Google ScholarĀ 

  48. Ferrarese C, Aliprandi A, Tremolizzo L, Stanzani L, De Micheli A, Dolara A, Frattola L. Increased glutamate in CSF and plasma of patients with HIV dementia. J Neuro-Oncol. 2001;57:671ā€“5.

    CASĀ  Google ScholarĀ 

  49. Benos DJ, Hahnt BH, Bubient JK, Ghosht SK, Mashburn NA, Chaikin MA, et al. Envelope glycoprotein gpl20 of human immunodeficiency. Proc Natl Acad Sci. 1994;91:494ā€“8.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  50. Sitcheran R, Gupta P, Fisher PB, Baldwin AS. Positive and negative regulation of EAAT2 by NF-kappaB: a role for N-myc in TNFalpha-controlled repression. EMBO J. 2005;24:510ā€“20.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  51. Schifitto G, Yiannoutsos CT, Simpson DM, Marra CM, Singer EJ, Kolson DL, et al. A placebo-controlled study of memantine for the treatment of human immunodeficiency virus-associated sensory neuropathy. J Neuro-Oncol. 2006;12:328ā€“31.

    CASĀ  Google ScholarĀ 

  52. Erdmann N, Zhao J, Lopez AL, Herek S, Curthoys N, Hexum TD, et al. Glutamate production by HIV-1 infected human macrophage is blocked by the inhibition of glutaminase. J Neurochem. 2007;102:539ā€“49.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  53. Thomas AG, Sattler R, Tendyke K, Loiacono KA, Hansen H, Sahni V, et al. High-throughput assay development for cystine-glutamate antiporter (xc-) highlights faster cystine uptake than glutamate release in glioma cells. PLoS One. 2015;10:1ā€“20.

    Google ScholarĀ 

  54. Ching J, Amiridis S, Stylli SS, Bjorksten AR, Kountouri N, Zheng T, et al. The peroxisome proliferator activated receptor gamma agonist pioglitazone increases functional expression of the glutamate transporter excitatory amino acid transporter 2 (EAAT2) in human glioblastoma cells. Oncotarget. 2015;6:21301ā€“14.

    ArticleĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  55. Dentesano G, Serratosa J, Tusell JM, RamĆ³n P, Valente T, Saura J, et al. CD200R1 and CD200 expression are regulated by PPAR-gamma in activated glial cells. Glia. 2014;62:982ā€“98.

    ArticleĀ  PubMedĀ  Google ScholarĀ 

  56. Li H, Singh S, Potula R, Persidsky Y, Kanmogne GD. Dysregulation of claudin-5 in HIV-induced interstitial pneumonitis and lung vascular injury: protective role of peroxisome proliferator-activated receptor-gamma. Am J Respir Crit Care Med. 2014;190:85ā€“97.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  57. Lakshmi SP, Reddy AT, Zhang Y, Sciurba FC, Mallampalli RK, Duncan SR, et al. Down-regulated peroxisome proliferator-activated receptor-gamma (PPARgamma) in lung epithelial cells promotes a PPARgamma agonist-reversible proinflammatory phenotype in chronic obstructive pulmonary disease (COPD). J Biol Chem. 2014;289:6383ā€“93.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  58. Zingarelli B, Sheehan M, Hake PW, O'Connor M, Denenberg A, Cook JA. Peroxisome proliferator-activated receptor-gamma ligands, 15-deoxyāˆ†12,14-prostaglandin J2 and ciglitazone, reduce systemic inflammation in polymicrobial sepsis by modulation of signal transduction pathways. J Immunol. 2003;171:6827ā€“37.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  59. Hoesel B, Schmid JA. The complexity of NF-ĪŗB signaling in inflammation and cancer. Mol Cancer. 2013;12:86.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  60. Hiscott J, Kwon H, GĆ©nin P. Hostile takeovers: viral appropriation of the NF-ĪŗB pathway. J Clin Invest. 2001;107:143ā€“51.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

Download references

Acknowledgements

The authors thank Ms. Amy Kao for initial assistance with the animal work. Dr. Reina Bendayan is a career scientist of Ontario HIV Treatment Network (OHTN), Ministry of Health Ontario. Amila Omeragic is the recipient of the Ontario Graduate Scholarship and the Leslie Dan Faculty of Pharmacy Deanā€™s Scholarship.

Funding

The work from this manuscript is supported by the Leslie Dan Faculty of Pharmacy Internal Fund allocated to Dr. Reina Bendayan.

Availability of data and materials

The datasets used and or analyzed during the current study are available from the corresponding author on a reasonable request.

Author information

Authors and Affiliations

Authors

Contributions

AO and RB designed the study. AO, TH, and UC performed the experiments. AO and RB contributed to the manuscript preparation. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Reina Bendayan.

Ethics declarations

Ethics approval

All procedures were carried out in accordance with the approval of the University of Toronto Animal Care Committee.

Consent for publication

Not Applicable.

Competing interests

The authors declare that they have no competing interests.

Publisherā€™s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Additional files

Additional file 1: Figure S1.

Effect of DMSO, HIV-1ADA gp120, PPARĪ³ agonists on cell viability in vitro. Primary cultures of mixed rat astrocytes and microglia were treated with either DMSO, gp120 (5Ā nM), rosiglitazone (1Ā Ī¼M), pioglitazone (20Ā Ī¼M, 50Ā Ī¼M) or GW9662 (500Ā nM) for 6Ā h, and cell viability was assessed using MTT assay. Results are expressed as percent of control and reported as meanĀ Ā±Ā SEM of at least 3 separate experiments (PDF 25 kb)

Additional file 2: Figure S2.

PPARĪ³ agonists rosiglitazone and pioglitazone reverse HIV-1ADA gp120-mediated inflammatory responses in vitro. Primary cultures of mixed rat astrocytes and microglia were treated with PPARĪ³ agonists, rosiglitazone (250Ā nMā€“1Ā Ī¼M) or pioglitazone (1ā€“50Ā Ī¼M) for 1Ā h prior to gp120 (5Ā nM) exposure for 3Ā h and. (A) TNF-Ī±, (B) IL-1Ī², and (C) iNOS mRNA levels were measured using qPCR. Cyclophilin BĀ was used as the housekeeping gene. Results are expressed as meanĀ Ā±Ā SEM relative to DMSO of at least 3 separate experiments. Asterisks and pound symbol represent data points significantly different from DMSO (control) and gp120 (vehicle) respectively (*pĀ <Ā 0.05, **pĀ <Ā 0.01, #pĀ <Ā 0.05, ##pĀ <Ā 0.01) (A-C) (PDF 47 kb)

Additional file 3: Figure S3.

PPARĪ³ agonist rosiglitazone reverses HIV-1ADA gp120-mediated inflammatory responses in frontal cortex. Adult Wistar rats were administered IP, 30Ā min prior to ICV bilateral injection of 4Ā Ī¼g/ventricle HIV-1ADA gp120 with rosiglitazone (10Ā mg/kg) or co-administration of rosiglitazone with GW9662 (5Ā mg/kg). Saline (control) and gp120 (vehicle) animals received the same volume of DMSO/saline 1:10 IP. Frontal cortex brain regions were isolated 24Ā h post ICV and (A) TNFĪ± and (B) IL-1Ī² and indicator of oxidative stress response (C) iNOS mRNA levels were measured using qPCR. Cyclophillin was used as the housekeeping gene. Results are expressed as meanĀ Ā±Ā SEM relative to saline group (control) nĀ =Ā 7ā€“12 animals/group. Asterisks and pound symbol represent data points significantly different from saline (control), and gp120 (vehicle) respectively. (*pĀ <Ā 0.05, **pĀ <Ā 0.01, #pĀ <Ā 0.05, ####pĀ <Ā 0.0001) (PDF 67 kb)

Additional file 4: Figure S4.

Effect of HIV-1ADA gp120 on the mRNA and protein expression of PPARĪ³ in frontal cortex. Adult Wistar rats were administered, bilateral ICV, 4Ā Ī¼g/ventricle of gp120, brain tissue was isolated 6ā€“72Ā h post ICV. PPARĪ³ mRNA expression was measured using qPCR. Cyclophillin was used as the housekeeping gene. For immunoblotting, frontal cortex tissue protein lysates (50Ā Ī¼g) were resolved on a 10% SDS-polyacrylamide gel and transferred to a PVDF membrane. HepG2 (50Ā Ī¼g) cells were used as positive control for PPARĪ³ protein. PPARĪ³ was detected using a rabbit polyclonal PPARĪ³ antibody (1:1000 dilution). Actin was detected using a mouse monoclonal antibody (1:5000, dilution). Data generated from densitometric analysis is presented as a ratio of PPARĪ³ expression normalized to actin (loading control). Results are expressed as meanĀ Ā±Ā SEM relative to saline group (control) nĀ =Ā 4ā€“12 animals/group. Asterisks represent data point significantly different from saline (control) animals (*pĀ <Ā 0.05, **pĀ <Ā 0.01) (PDF 94 kb)

Rights and permissions

Open Access This 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.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Omeragic, A., Hoque, M., Choi, Uy. et al. Peroxisome proliferator-activated receptor-gamma: potential molecular therapeutic target for HIV-1-associated brain inflammation. J Neuroinflammation 14, 183 (2017). https://doi.org/10.1186/s12974-017-0957-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12974-017-0957-8

Keywords