Injection drug use increases the probability of contracting HIV, and opioids accelerate the progression of HIV-1 infection through immune suppression and direct CNS actions. Opioid receptors are widely expressed on immune cells and glia, and opioids can modulate chemokine signaling. CCR5 plays a critical role in HIV infection as a co-receptor for entry of CCR5-preferring strains. CCR5 levels in general, and in microglia/CNS macrophages, have been correlated with severity of HIV neurologic disease. Individuals homozygous for the CCR5?32 mutation resist infection, and the CCR5 antagonist maraviroc (MVC) is used clinically to inhibit HIV entry. Results from the last funding period using non-infective paradigms reveal that CCR5 may also promote HIV-related neuropathology apart from its role in enhancing HIV infection. For example, blockade of CCR5 with MVC increases synaptic plasticity, reverses Tat-induced reductions in antinocicetion, tolerance, and morphine dependence, and enhances survival of Tat-treated striatal neurons. CCR5 overactivation may be a pivotal factor in the ability of opiates to amplify HIV neuropathogenesis, and heterologous interactions between MOR and CCR5 likely underlie many CNS deficits in HAND. CCR5 and MOR interactions in the context of HIV are likely fundamentally different among CNS regions due to distinct glial/neuronal distributions of CCR5 and MOR, their ligands, and effector coupling. Critical to this proposal, MVC rescued Tat-induced deficits in a spatial learning/memory task involving hippocampal function (Barnes maze), and hippocampal damage is involved in learning/memory deficits in HAND patients, irrespective of cART. To explore the hypothesis that aberrant CCR5-MOR interactions underlie HIV-induced hippocampal deficits, we will systematically interrogate those interactions in the context of HIV/Tat and opiate exposure. In vivo and ex vivo studies in HIV-1 Tat transgenic mice are complemented by in vitro studies using human neurons exposed to Tat, gp120, and HIV. Three interrelated Aims test hypotheses in male and female mice.
Aim 1 tests CCR5 impact on the dynamics of HIV-1/Tat and MOR interactions in the hippocampus, using functional assessments of behavior (spatial and non-spatial tasks) and neurophysiology (LTP and membrane properties in ex vivo slices).
Aim 2 tests how CCR5 activation or blockade (genetic, MVC) alters HIV-1/Tat and MOR interactions, using Tat, gp120, and HIV in murine or human co-cultures of hippocampal neurons and glia. Neuron dysfunction (optical physiology), [Ca2+]i homeostasis, synaptodendritic injury, and death, as well as underlying inflammation and signaling pathways involved (bioplex/MSD multiplex) are explored.
Aim 3 examines heterologous interactions between MOR and CCR5, assessing receptor dynamics and activity by [3H]naloxone or [3H]maraviroc-stimulated [35S]GTP?S autoradiography in hippocampal sections, and concentration-effect binding curves in membranes from brain and cultured neurons/glia. TrkB and p75NTR roles in regulating outcomes tested pharmacologically; protection by altering the proBDNF/mBDNF balance (tPA to increase proBDNF cleavage) assessed in all Aims.

Public Health Relevance

Opioid drug abuse appears to enhance CNS deficits that occur due to HIV-1 infection. In the past funding period we established that signals through -opioid and CCR5 receptors uniquely interact to promote inflammation, neuronal injury/death, and dysfunction in the striatum, and we developed new preliminary data that CCR5 blockade with maraviroc reverses performance deficits in a spatial memory test (Barnes maze) reflecting hippocampal function. Since cART-treated HIV patients still show hippocampal damage and have related cognitive deficits, and since the cellular expression of CCR5 and -opioid receptors and their ligands and signaling partners are unique to different brain regions, we will examine the unique, interactive role of MOR and CCR5 in orchestrating synergistic deficits in hippocampal pathology, neuronal dysfunction, and learning and memory, and assesses whether drug therapies that either inhibit CCR5 directly (maraviroc), or via the BDNF pathway (BDNF, tPA) will reverse detrimental effects of opiate drugs and HIV, using in vivo (behavior, structure), ex vivo (electrophysiology), and in vitro manipulations of neurons, glia, and brain aggregates (human primary and iPSC-derived).

Agency
National Institute of Health (NIH)
Institute
National Institute on Drug Abuse (NIDA)
Type
Research Project (R01)
Project #
5R01DA034231-07
Application #
9937696
Study Section
Special Emphasis Panel (ZRG1)
Program Officer
Lin, Yu
Project Start
2013-04-15
Project End
2024-03-31
Budget Start
2020-04-01
Budget End
2021-03-31
Support Year
7
Fiscal Year
2020
Total Cost
Indirect Cost
Name
Virginia Commonwealth University
Department
Anatomy/Cell Biology
Type
Schools of Medicine
DUNS #
105300446
City
Richmond
State
VA
Country
United States
Zip Code
23298
Kim, Sarah; Hahn, Yun Kyung; Podhaizer, Elizabeth M et al. (2018) A central role for glial CCR5 in directing the neuropathological interactions of HIV-1 Tat and opiates. J Neuroinflammation 15:285
Wodarski, Rachel; Bagdas, Deniz; Paris, Jason J et al. (2018) Reduced intraepidermal nerve fibre density, glial activation, and sensory changes in HIV type-1 Tat-expressing female mice: involvement of Tat during early stages of HIV-associated painful sensory neuropathy. Pain Rep 3:e654
Gonek, Maciej; McLane, Virginia D; Stevens, David L et al. (2018) CCR5 mediates HIV-1 Tat-induced neuroinflammation and influences morphine tolerance, dependence, and reward. Brain Behav Immun 69:124-138
Leibrand, Crystal R; Paris, Jason J; Ghandour, M Said et al. (2017) HIV-1 Tat disrupts blood-brain barrier integrity and increases phagocytic perivascular macrophages and microglia in the dorsal striatum of transgenic mice. Neurosci Lett 640:136-143
Schier, Christina J; Marks, William D; Paris, Jason J et al. (2017) Selective Vulnerability of Striatal D2 versus D1 Dopamine Receptor-Expressing Medium Spiny Neurons in HIV-1 Tat Transgenic Male Mice. J Neurosci 37:5758-5769
Poland, Ryan S; Hahn, Yun; Knapp, Pamela E et al. (2016) Ibudilast attenuates expression of behavioral sensitization to cocaine in male and female rats. Neuropharmacology 109:281-292
Marks, William D; Paris, Jason J; Schier, Christina J et al. (2016) HIV-1 Tat causes cognitive deficits and selective loss of parvalbumin, somatostatin, and neuronal nitric oxide synthase expressing hippocampal CA1 interneuron subpopulations. J Neurovirol 22:747-762
Arnatt, Christopher K; Falls, Bethany A; Yuan, Yunyun et al. (2016) Exploration of bivalent ligands targeting putative mu opioid receptor and chemokine receptor CCR5 dimerization. Bioorg Med Chem 24:5969-5987
Fitting, Sylvia; Stevens, David L; Khan, Fayez A et al. (2016) Morphine Tolerance and Physical Dependence Are Altered in Conditional HIV-1 Tat Transgenic Mice. J Pharmacol Exp Ther 356:96-105
Hahn, Yun K; Paris, Jason J; Lichtman, Aron H et al. (2016) Central HIV-1 Tat exposure elevates anxiety and fear conditioned responses of male mice concurrent with altered mu-opioid receptor-mediated G-protein activation and ?-arrestin 2 activity in the forebrain. Neurobiol Dis 92:124-36

Showing the most recent 10 out of 19 publications