Abstract

BackgroundDespite 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 PPARγ exerts anti-inflammatory properties in neurological disorders. The goal of this study was to examine the role of PPARγ 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.MethodsPrimary mixed cultures of rat astrocytes and microglia were treated with PPARγ agonists (rosiglitazone or pioglitazone) and exposed to HIV-1ADA gp120. Inflammatory cytokines and indicator of oxidative stress response (TNFα, IL-1β, 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 PPARγ agonist (rosiglitazone) or co-administration with PPARγ antagonist (GW9662). qPCR and immunoblotting analyses were applied to measure inflammatory markers, GLT-1 and PPARγ.ResultsIn 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 PPARγ agonist rosiglitazone were mediated through inhibition of NF-κB.ConclusionOur 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 PPARγ agonists, rosiglitazone or pioglitazone. Together our data suggest that targeting PPARγ signaling may provide an option for preventing/treating HIV-associated brain inflammation.

Highlights

  • Despite the use of combination antiretroviral therapy for the treatment of Human immunodeficiency virus (HIV)-1 infection, cognitive impairments remain prevalent due to persistent viral replication and associated brain inflammation

  • We have successfully shown that these effects can be reversed by treatment with Peroxisome proliferator-activated receptor gamma (PPARγ) agonists, rosiglitazone or pioglitazone

  • Together our data suggest that targeting PPARγ signaling may provide an option for preventing/treating HIV-associated brain inflammation

Read more

Summary

Introduction

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. In response to HIV-1, microglia and astrocytes become activated and secrete proinflammatory cytokines [i.e., tumor necrosis factor-α (TNFα), interleukin-1β (IL-1β), 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]. 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 gp120associated brain inflammation [6,7,8]

Objectives
Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call