28 years after the beginning of the AIDS epidemic, we are confronting the fact that more than one out of three people with HIV-1 develop some form of HIV-1 associated neurocognitive disorder (HAND). Highly active antiretroviral therapy (HAART), even with dramatic reduction of viral load, may reverse some symptoms of HAND (often on a transient basis), but neither cures or prevents the disease. This RO1 has investigated the role of the phospholipid mediator platelet activating factor (PAF) in normal and pathologic synaptic transmission as a key molecule involved in the neuropathogenesis of HAND. In this competing renewal, we hypothesize that viral proteins from HIV-1 can disrupt normal PAF signaling between neuronal synapses by direct and enduring effects on CNS-trafficking immune effector cells that amplify the effects of PAF and cause neurologic disease. Building on data from the previous cycle of this RO1, prolonged exposure or increasing doses of PAF results in a continuum of reversible to irreversible synaptodendritic damage with failure of synaptic transmission due to mitochondrial dysfunction. In this application, we will use in vivo multiphoton microscopy (MPM) to investigate synaptodendritic damage during conditions that model HAND in mouse cortex, using transgenic animals with fluorescently labeled neurons and/or mononuclear phagocytes. We propose three specific aims using HIV-1 Tat and its downstream effector, PAF, to induce changes in immune cell trafficking into the CNS and synaptic communication.
In aim 1, we will characterize in vivo PAF-induced dendritic beading that may become irreversible after PAF concentrations exceed thresholds for excitotoxic and neuroinflammatory damage, with the goal of understanding how increasing doses of PAF affect synaptic architecture and function during neuroinflammatory conditions that model HAND.
In aim 2, we will use immunodepletion techniques to selectively remove peripheral mononuclear cells or neutrophils prior to stereotactic injection of HIV-1 Tat into the brain, to test whether these cells contribute to synaptodendritic damage by phagocytic engulfment or release of soluble mediators.
In aim 3, we will determine the in vivo kinetics of cortical NADH production in response to Tat and PAF, as a measure of tissue oxygenation and energy metabolism;these experiments will test whether synaptodendritic damage occurs due to energy failure in response to increased synaptic activity during neuroinflammation. Together, these aims will help us translate our model of HIV-1 neuropathogenesis to in vivo conditions, bringing us closer to an understanding of how HAND continues to be a problem of major importance to people living with HIV-1.

Public Health Relevance

This is the fourth cycle of a research application that seeks to understand the role of a signaling molecule, platelet activating factor (PAF), normally produced from cellular membranes in immune cells and neurons, in the disease state caused by human immunodeficiency virus type 1 (HIV-1) infection in the brain. In our previous cycles we have defined a relationship between the HIV-1 regulatory protein, Tat, and the production of PAF, that increases levels of PAF in the brain and disrupts normal cellular signaling between neurons at the synapse, a specialized portion of two neurons that is necessary to generate the electrical and chemical signals necessary for communication in the brain. The major purpose of this cycle of the grant is to understand how these events occur in the brain of mice that are genetically engineered to help us visualize the synapse. This in turn will help us gain greater insight into how HIV-1 continues to cause neurologic disease despite effective control of its life cycle by highly active antiretroviral therapy (HAART).

Agency
National Institute of Health (NIH)
Institute
National Institute of Mental Health (NIMH)
Type
Research Project (R01)
Project #
5R01MH056838-15
Application #
7993046
Study Section
Special Emphasis Panel (ZRG1-AARR-D (02))
Program Officer
Joseph, Jeymohan
Project Start
1996-09-30
Project End
2014-12-31
Budget Start
2011-01-01
Budget End
2011-12-31
Support Year
15
Fiscal Year
2011
Total Cost
$362,093
Indirect Cost
Name
University of Rochester
Department
Neurology
Type
Schools of Dentistry
DUNS #
041294109
City
Rochester
State
NY
Country
United States
Zip Code
14627
Bellizzi, Matthew J; Geathers, Jasmine S; Allan, Kevin C et al. (2016) Platelet-Activating Factor Receptors Mediate Excitatory Postsynaptic Hippocampal Injury in Experimental Autoimmune Encephalomyelitis. J Neurosci 36:1336-46
Zhang, Gang; Guo, Dongwei; Dash, Prasanta K et al. (2016) The mixed lineage kinase-3 inhibitor URMC-099 improves therapeutic outcomes for long-acting antiretroviral therapy. Nanomedicine 12:109-22
Puccini, Jenna M; Marker, Daniel F; Fitzgerald, Tim et al. (2015) Leucine-rich repeat kinase 2 modulates neuroinflammation and neurotoxicity in models of human immunodeficiency virus 1-associated neurocognitive disorders. J Neurosci 35:5271-83
Tremblay, Marie-Ève; Marker, Daniel F; Puccini, Jenna M et al. (2013) Ultrastructure of microglia-synapse interactions in the HIV-1 Tat-injected murine central nervous system. Commun Integr Biol 6:e27670
Gelbard, Harris A; Gendelman, Howard E (2013) Lipids and cognition make good bedfellows for neuroAIDS. Neurology 81:1480-1
Marker, Daniel F; Tremblay, Marie-Ève; Puccini, Jenna M et al. (2013) The new small-molecule mixed-lineage kinase 3 inhibitor URMC-099 is neuroprotective and anti-inflammatory in models of human immunodeficiency virus-associated neurocognitive disorders. J Neurosci 33:9998-10010
Marker, Daniel F; Puccini, Jenna M; Mockus, Taryn E et al. (2012) LRRK2 kinase inhibition prevents pathological microglial phagocytosis in response to HIV-1 Tat protein. J Neuroinflammation 9:261
Lu, Shao-Ming; Tremblay, Marie-Ève; King, Irah L et al. (2011) HIV-1 Tat-induced microgliosis and synaptic damage via interactions between peripheral and central myeloid cells. PLoS One 6:e23915
Perry, Seth W; Norman, John P; Barbieri, Justin et al. (2011) Mitochondrial membrane potential probes and the proton gradient: a practical usage guide. Biotechniques 50:98-115
Ramirez, Servio H; Fan, Shongshan; Dykstra, Holly et al. (2010) Dyad of CD40/CD40 ligand fosters neuroinflammation at the blood-brain barrier and is regulated via JNK signaling: implications for HIV-1 encephalitis. J Neurosci 30:9454-64

Showing the most recent 10 out of 38 publications