Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
J Neurovirol ; 30(1): 1-21, 2024 02.
Article in English | MEDLINE | ID: mdl-38280928

ABSTRACT

Opioid overdose deaths have dramatically increased by 781% from 1999 to 2021. In the setting of HIV, opioid drug abuse exacerbates neurotoxic effects of HIV in the brain, as opioids enhance viral replication, promote neuronal dysfunction and injury, and dysregulate an already compromised inflammatory response. Despite the rise in fentanyl abuse and the close association between opioid abuse and HIV infection, the interactive comorbidity between fentanyl abuse and HIV has yet to be examined in vivo. The HIV-1 Tat-transgenic mouse model was used to understand the interactive effects between fentanyl and HIV. Tat is an essential protein produced during HIV that drives the transcription of new virions and exerts neurotoxic effects within the brain. The Tat-transgenic mouse model uses a glial fibrillary acidic protein (GFAP)-driven tetracycline promoter which limits Tat production to the brain and this model is well used for examining mechanisms related to neuroHIV. After 7 days of fentanyl exposure, brains were harvested. Tight junction proteins, the vascular cell adhesion molecule, and platelet-derived growth factor receptor-ß were measured to examine the integrity of the blood brain barrier. The immune response was assessed using a mouse-specific multiplex chemokine assay. For the first time in vivo, we demonstrate that fentanyl by itself can severely disrupt the blood-brain barrier and dysregulate the immune response. In addition, we reveal associations between inflammatory markers and tight junction proteins at the blood-brain barrier.


Subject(s)
Blood-Brain Barrier , Fentanyl , HIV-1 , Mice, Transgenic , Neuroinflammatory Diseases , tat Gene Products, Human Immunodeficiency Virus , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Blood-Brain Barrier/virology , Mice , Fentanyl/pharmacology , HIV-1/drug effects , HIV-1/genetics , tat Gene Products, Human Immunodeficiency Virus/genetics , tat Gene Products, Human Immunodeficiency Virus/metabolism , Neuroinflammatory Diseases/genetics , Neuroinflammatory Diseases/pathology , Neuroinflammatory Diseases/virology , HIV Infections/virology , HIV Infections/genetics , HIV Infections/pathology , HIV Infections/drug therapy , Disease Models, Animal , Analgesics, Opioid/pharmacology , Analgesics, Opioid/adverse effects , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Tight Junction Proteins/metabolism , Tight Junction Proteins/genetics , Humans , Brain/drug effects , Brain/virology , Brain/metabolism , Brain/pathology , Opioid-Related Disorders/genetics , Opioid-Related Disorders/pathology , Opioid-Related Disorders/metabolism
2.
Anal Bioanal Chem ; 415(23): 5809-5817, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37490153

ABSTRACT

Here, we present a method developed for the analysis of spatial distributions of morphine in mouse brain tissue using infrared matrix-assisted laser desorption electrospray ionization (IR-MALDESI) coupled to a Q Exactive Plus mass spectrometer. The method is also capable of evaluating spatial distributions of the antiretroviral drug abacavir. To maximize sensitivity to morphine, we analyze various Orbitrap mass spectrometry acquisition modes utilizing signal abundance and frequency of detection as evaluation criteria. We demonstrate detection of morphine in mouse brain and establish that the selected ion monitoring mode provides 2.5 times higher sensitivity than the full-scan mode. We find that distributions of morphine and abacavir are highly correlated with the Pearson correlation coefficient R = 0.87. Calibration showed that instrument response is linear up to 40 pg/mm2 (3.8 µg/g of tissue).


Subject(s)
Morphine , Spectrometry, Mass, Electrospray Ionization , Mice , Animals , Spectrometry, Mass, Electrospray Ionization/methods , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Brain , Lasers
3.
Neurosci Lett ; 788: 136852, 2022 09 25.
Article in English | MEDLINE | ID: mdl-36028004

ABSTRACT

Despite advances in the treatment of human immunodeficiency virus (HIV), approximately one-half of people infected with HIV (PWH) experience neurocognitive impairment. Opioid use disorder (OUD) can exacerbate the cognitive and pathological changes seen in PWH. HIV increases inflammation and immune cell trafficking into the brain; however, less is known about how opioid use disorder affects the recruitment of immune cells. Accordingly, we examined the temporal consequences of HIV-1 Tat and/or morphine on the recruitment of endocytic cells (predominantly perivascular macrophages and microglia) in the dorsal striatum and hippocampus by infusing multi-colored, fluorescently labeled dextrans before and after exposure. To address this question, transgenic mice that conditionally expressed HIV-1 Tat (Tat+), or their control counterparts (Tat-), received three sequential intracerebroventricular (i.c.v.) infusions of Cascade Blue-, Alexa Fluor 488-, and Alexa Fluor 594-labeled dextrans, respectively infused 1 day before, 1-day after, or 13-days after morphine and/or Tat exposure. At the end of the study, the number of cells labeled with each fluorescent dextran were counted. The data demonstrated a significantly higher influx of newly-labeled cells into the perivascular space than into the parenchyma. In the striatum, Tat or morphine exposure increased the number of endocytic cells in the perivascular space, while only morphine increased the recruitment of endocytic cells into the parenchyma. In the hippocampus, morphine (but not Tat) increased the influx of dextran-labeled cells into the perivascular space, but there were too few labeled cells within the hippocampal parenchyma to analyze. Collectively, these data suggest that HIV-1 Tat and morphine act independently to increase the recruitment of endocytic cells into the brain in a region-specific manner.


Subject(s)
HIV Infections , HIV-1 , Opioid-Related Disorders , Animals , Corpus Striatum/metabolism , Dextrans , Fluoresceins , HIV-1/metabolism , Humans , Macrophages/metabolism , Mice , Mice, Transgenic , Morphine/pharmacology , Sulfonic Acids , tat Gene Products, Human Immunodeficiency Virus/metabolism
4.
J Neurovirol ; 25(4): 560-577, 2019 08.
Article in English | MEDLINE | ID: mdl-31102185

ABSTRACT

Poor antiretroviral penetration may contribute to human immunodeficiency virus (HIV) persistence within the brain and to neurocognitive deficits in opiate abusers. To investigate this problem, HIV-1 Tat protein and morphine effects on blood-brain barrier (BBB) permeability and drug brain penetration were explored using a conditional HIV-1 Tat transgenic mouse model. Tat and morphine effects on the leakage of fluorescently labeled dextrans (10-, 40-, and 70-kDa) into the brain were assessed. To evaluate effects on antiretroviral brain penetration, Tat+ and Tat- mice received three antiretroviral drugs (dolutegravir, abacavir, and lamivudine) with or without concurrent morphine exposure. Antiretroviral and morphine brain and plasma concentrations were determined by LC-MS/MS. Morphine exposure, and, to a lesser extent, Tat, significantly increased tracer leakage from the vasculature into the brain. Despite enhanced BBB breakdown evidenced by increased tracer leakiness, morphine exposure led to significantly lower abacavir concentrations within the striatum and significantly less dolutegravir within the hippocampus and striatum (normalized to plasma). P-glycoprotein, an efflux transporter for which these drugs are substrates, expression and function were significantly increased in the brains of morphine-exposed mice compared to mice not exposed to morphine. These findings were consistent with lower antiretroviral concentrations in brain tissues examined. Lamivudine concentrations were unaffected by Tat or morphine exposure. Collectively, our investigations indicate that Tat and morphine differentially alter BBB integrity. Morphine decreased brain concentrations of specific antiretroviral drugs, perhaps via increased expression of the drug efflux transporter, P-glycoprotein.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Blood-Brain Barrier/drug effects , HIV-1/genetics , Morphine/adverse effects , tat Gene Products, Human Immunodeficiency Virus/biosynthesis , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Biological Transport/drug effects , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/virology , Capillary Permeability , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Corpus Striatum/virology , Dextrans/pharmacokinetics , Dideoxynucleosides/pharmacokinetics , Female , Fluorescein-5-isothiocyanate/analogs & derivatives , Fluorescein-5-isothiocyanate/pharmacokinetics , HIV Infections/metabolism , HIV Infections/psychology , HIV Infections/virology , HIV-1/metabolism , Heterocyclic Compounds, 3-Ring/pharmacokinetics , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/virology , Lamivudine/pharmacokinetics , Mice , Mice, Transgenic , Models, Biological , Neurocognitive Disorders/metabolism , Neurocognitive Disorders/psychology , Neurocognitive Disorders/virology , Oxazines , Piperazines , Pyridones , tat Gene Products, Human Immunodeficiency Virus/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...