Methamphetamine (Meth) abuse leads to behaviors that increase the risk of exposure to HIV1-3. NeuroAIDS, which affects >40% of HIV+ individuals, is a particularly severe complication of the infection causing neurological deficits, not decreased by antiviral drugs that do not cross the blood-brain-barrier efficiently 4. NeuroAIDS and peripheral pathologies associated to this co-morbidity have been reported to be aggravated by Meth5 6. Thus, the effects of Meth are predominant, but not restricted to the CNS. We have found that Meth modulates the phenotype of brain and peripheral innate immune cells in the SIV/Rhesus macaque model, increasing cytokine secretion and enriching virus targets CCR5+ cells7, and in vitro. We will investigate the hypothesis of direct action of Meth on innate immune cells, causing alterations that may enhance virus-triggered tissue-pathologies in HIV+ Meth abusers. In the present proposal, we will focus on innate immune cells existing in the brain and in the periphery, especially macrophages and microglia cells. Given that, we hypothesize that Meth can interfere with the anti-viral host immune response, by directly affecting innate immune cells of the macrophage/ microglia lineage. This can be an important component of increased neuropathology, particularly in neuroAIDS, since HIV brain targets are macrophages and microglia. We will determine if Meth modulates gene expression in CNS-derived immune cells in SIV-infected macaques, and changes sets of genes triggered by SIV in the brain by comparing gene expression in spleen and brain- cryopreserved immune cells (depleted of neurons and astro/oligoglia) in healthy and SIV-infected macaques, treated or not with Meth. Expression data will be compared using an integrative systems biology approach and validated. We will prioritize high score pathways, but favor molecules involved in cell cycle, oxidative stress, and inflammatory phenotype, in which the effects of Meth have been observed. Validation will be performed using qRTPCR and protein detection (western blot on brain and spleen frozen tissue fragments, FACS of frozen cell suspensions, and immunohistochemistry on paraffin-embedded tissue - already processed and available for all animals). In parallel, we will assess the ability of Meth to directly modulate macrophage/ microglia gene/protein expression and functional characteristics, using macrophage cell lines and human microglia primary cultures. Gene expression and functional assays assess the effects of Meth on cell cycle, oxidative stress, chemokine receptor and inflammatory molecule expression, mitochondrial functions and antigen processing/ presentation. This will allow the observation of direct effects on innate immune cells that get infected and predominate in the brain. These studies will help understand mechanisms regulating the severity of brain and other tissues pathology in HIV+ Meth users and reveal direct Meth targets on innate immune cells.

Public Health Relevance

Among Methamphetamine (Meth) users, there is a large overlap with HIV infection, being NeuroAIDS an important complication reported to be aggravated by this drug, although its effects on pathology are not restricted to the CNS. Given that the mechanisms by which the drug aggravates pathology and operates systemically are not well understood, there are no medications for treating the pathological consequences of Meth use in HIV or in other viral infections. To help understand mechanisms regulating the severity of brain and other tissue pathologies in HIV+ Meth, the present exploratory study will investigate direct effects on innate immune cells, which are predominantly infected in the brain and present also in other inflammatory conditions, aiming a molecular basis for therapeutic intervention, and for a new paradigm of immune-mediated pathology in drug abuse.

Agency
National Institute of Health (NIH)
Institute
National Institute on Drug Abuse (NIDA)
Type
Exploratory/Developmental Grants (R21)
Project #
1R21DA029491-01A1
Application #
8067738
Study Section
NeuroAIDS and other End-Organ Diseases Study Section (NAED)
Program Officer
Avila, Albert
Project Start
2011-09-01
Project End
2013-08-31
Budget Start
2011-09-01
Budget End
2012-08-31
Support Year
1
Fiscal Year
2011
Total Cost
$284,250
Indirect Cost
Name
Scripps Research Institute
Department
Type
DUNS #
781613492
City
La Jolla
State
CA
Country
United States
Zip Code
92037
Najera, Julia A; Bustamante, Eduardo A; Bortell, Nikki et al. (2016) Methamphetamine abuse affects gene expression in brain-derived microglia of SIV-infected macaques to enhance inflammation and promote virus targets. BMC Immunol 17:7
Bortell, Nikki; Morsey, Brenda; Basova, Liana et al. (2015) Phenotypic changes in the brain of SIV-infected macaques exposed to methamphetamine parallel macrophage activation patterns induced by the common gamma-chain cytokine system. Front Microbiol 6:900
Marcondes, Maria Cecilia G; Morsey, Brenda; Emanuel, Katy et al. (2015) CD8+ T cells maintain suppression of simian immunodeficiency virus in the central nervous system. J Infect Dis 211:40-4
Marcondes, Maria Cecilia Garibaldi; Ojakian, Ryan; Bortell, Nikki et al. (2014) Osteopontin expression in the brain triggers localized inflammation and cell death when immune cells are activated by pertussis toxin. Mediators Inflamm 2014:358218
Sabouri, Amir H; Marcondes, Maria Cecilia Garibaldi; Flynn, Claudia et al. (2014) TLR signaling controls lethal encephalitis in WNV-infected brain. Brain Res 1574:84-95
Sanchez-Alavez, Manuel; Bortell, Nikki; Galmozzi, Andrea et al. (2014) Reactive oxygen species scavenger N-acetyl cysteine reduces methamphetamine-induced hyperthermia without affecting motor activity in mice. Temperature (Austin) 1:227-241
Sanchez-Alavez, Manuel; Conti, Bruno; Wood, Malcolm R et al. (2013) ROS and Sympathetically Mediated Mitochondria Activation in Brown Adipose Tissue Contribute to Methamphetamine-Induced Hyperthermia. Front Endocrinol (Lausanne) 4:44
Chaudhuri, Amrita Datta; Yelamanchili, Sowmya V; Marcondes, Maria Cecilia G et al. (2013) Up-regulation of microRNA-142 in simian immunodeficiency virus encephalitis leads to repression of sirtuin1. FASEB J 27:3720-9
Marcondes, Maria Cecilia Garibaldi; Spina, Celsa; Bustamante, Eduardo et al. (2013) Increased toll-like receptor signaling pathways characterize CD8+ cells in rapidly progressive SIV infection. Biomed Res Int 2013:796014
Alboni, Silvia; Montanari, Claudia; Benatti, Cristina et al. (2011) Constitutive and LPS-regulated expression of interleukin-18 receptor beta variants in the mouse brain. Brain Behav Immun 25:483-93