1
|
Kaufman MJ, Meloni EG. Xenon gas as a potential treatment for opioid use disorder, alcohol use disorder, and related disorders. Med Gas Res 2025; 15:234-253. [PMID: 39812023 PMCID: PMC11918480 DOI: 10.4103/mgr.medgasres-d-24-00063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 09/26/2024] [Indexed: 01/16/2025] Open
Abstract
Xenon gas is considered to be a safe anesthetic and imaging agent. Research on its other potentially beneficial effects suggests that xenon may have broad efficacy for treating health disorders. A number of reviews on xenon applications have been published, but none have focused on substance use disorders. Accordingly, we review xenon effects and targets relevant to the treatment of substance use disorders, with a focus on opioid use disorder and alcohol use disorder. We report that xenon inhaled at subsedative concentrations inhibits conditioned memory reconsolidation and opioid withdrawal symptoms. We review work by others reporting on the antidepressant, anxiolytic, and analgesic properties of xenon, which could diminish negative affective states and pain. We discuss research supporting the possibility that xenon could prevent analgesic- or stress-induced opioid tolerance and, by so doing could reduce the risk of developing opioid use disorder. The rapid kinetics, favorable safety and side effect profiles, and multitargeting capability of xenon suggest that it could be used as an ambulatory on-demand treatment to rapidly attenuate maladaptive memory, physical and affective withdrawal symptoms, and pain drivers of substance use disorders when they occur. Xenon may also have human immunodeficiency virus and oncology applications because its effects relevant to substance use disorders could be exploited to target human immunodeficiency virus reservoirs, human immunodeficiency virus protein-induced abnormalities, and cancers. Although xenon is expensive, low concentrations exert beneficial effects, and gas separation, recovery, and recycling advancements will lower xenon costs, increasing the economic feasibility of its therapeutic use. More research is needed to better understand the remarkable repertoire of effects of xenon and its potential therapeutic applications.
Collapse
Affiliation(s)
- Marc J Kaufman
- McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | | |
Collapse
|
2
|
Xie Q, Dasari R, Namba MD, Buck LA, Side CM, Park K, Jackson JG, Barker JM. Astrocytic regulation of cocaine locomotor sensitization in EcoHIV infected mice. Neuropharmacology 2025; 265:110245. [PMID: 39631679 DOI: 10.1016/j.neuropharm.2024.110245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/30/2024] [Accepted: 11/30/2024] [Indexed: 12/07/2024]
Abstract
Cocaine use disorder (CUD) is highly comorbid with HIV infection and worsens HIV outcomes. Preclinical research on the outcomes of HIV infection may yield crucial information on neurobehavioral changes resulting from chronic drug exposure in people living with HIV (PLWH). Repeated exposure to cocaine alters behavioral responses to cocaine. This includes development of cocaine locomotor sensitization - or increased locomotor responses to the same doses of cocaine - which depends on nucleus accumbens (NAc) neural plasticity. NAc astrocytes are key regulators of neural activity and plasticity, and their function can be impaired by cocaine exposure and HIV infection, thus implicating them as potential regulators of HIV-induced changes in behavioral response to cocaine. To characterize the effects of HIV infection on cocaine locomotor sensitization, we employed the EcoHIV mouse model in male and female mice to assess changes in locomotor responses after repeated cocaine (10 mg/kg) exposure and challenge. EcoHIV infection potentiated expression of cocaine sensitization. We also identified EcoHIV-induced increases in expression of the astrocytic nuclear marker Sox9 selectively in the NAc core. To investigate whether modulation of NAc astrocytes could reverse EcoHIV-induced deficits, we employed a chemogenetic approach. We found that chemogenetic activation of NAc astrocyte Gq signaling attenuated EcoHIV-enhanced cocaine sensitization. We propose that HIV infection contributes to cocaine behavioral sensitization and induces adaptations in NAc astrocytes, while promoting NAc astrocytic Gq-signaling can recover EcoHIV-induced behavioral changes. These findings identify potential cellular substrates of disordered cocaine-driven behavior in the context of HIV infection and point toward strategies to reduce cocaine-related behavior in PLWH.
Collapse
Affiliation(s)
- Qiaowei Xie
- Department of Pharmacology and Physiology, Drexel University College of Medicine, USA; Graduate Program in Pharmacology and Physiology, Drexel University College of Medicine, USA
| | - Rohan Dasari
- Department of Pharmacology and Physiology, Drexel University College of Medicine, USA
| | - Mark D Namba
- Department of Pharmacology and Physiology, Drexel University College of Medicine, USA
| | - Lauren A Buck
- Department of Pharmacology and Physiology, Drexel University College of Medicine, USA
| | - Christine M Side
- Department of Pharmacology and Physiology, Drexel University College of Medicine, USA
| | - Kyewon Park
- University of Pennsylvania Center for AIDS Research, USA
| | - Joshua G Jackson
- Department of Pharmacology and Physiology, Drexel University College of Medicine, USA
| | - Jacqueline M Barker
- Department of Pharmacology and Physiology, Drexel University College of Medicine, USA.
| |
Collapse
|
3
|
Jimenez-Torres AC, Hastie JA, Davis SE, Porter KD, Lei B, Moukha-Chafiq O, Zhang S, Nguyen TH, Ananthan S, Augelli-Szafran CE, Zhu J. Identification of pyrimidine structure-based compounds as allosteric ligands of the dopamine transporter as therapeutic agents for NeuroHIV. J Pharmacol Exp Ther 2025; 392:100021. [PMID: 40023582 DOI: 10.1124/jpet.124.002138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 05/14/2024] [Accepted: 07/23/2024] [Indexed: 01/22/2025] Open
Abstract
The disruption of dopamine (DA) neurotransmission by the HIV-1 transactivator of transcription (Tat) during HIV-1 infection has been linked to the development of neurocognitive disorders, even under combined antiretroviral therapy treatment. We have demonstrated that Southern Research Institute (SRI) 32742, a novel allosteric modulator of DA transporter (DAT), attenuates cocaine- and Tat-binding to DAT, alleviates Tat-induced cognitive deficits and potentiation of cocaine reward in inducible Tat transgenic mice. The current study determined the in vitro pharmacological profile of SRI-32743 and its optimized second-generation analogs and their effects as allosteric modulators. Through structure-activity relationship studies of SRI-32743, 170 compounds were synthesized and evaluated for their ability to modulate DAT function. We identified 21 analogs as atypical competitors of DAT (maximum attributable drug effect, ≤60%). Four compounds, SRI-46564, SRI-47056, SRI-46286, and SRI-47867, displayed IC50 values for [3H]DA uptake inhibition from 9.33 ± 0.50 to 0.96 ± 0.05 μM and from 3.96 ± 1.36 to 1.29 ± 0.19 for DAT binding, respectively. The 4 analogs also displayed high potency at 2 different concentrations (0.5 nM and 0.05 nM) to attenuate Tat-induced inhibition of [3H]DA uptake and cocaine-mediated dissociation of [3H]WIN35,428 binding in Chinese hamster ovary cells expressing human DAT, suggesting that the effects occur through an allosteric mechanism. In further ex vivo studies using fast scan cyclic voltammetry, we demonstrated that the analogs do not disrupt the baseline phasic-like DA release. These findings provide a new insight into the potential for development of novel therapeutic agents to attenuate DAT-Tat interactions to normalize DA neurotransmission in NeuroHIV. SIGNIFICANCE STATEMENT: The allosteric inhibition of the dopamine (DA) transporter by the HIV-1 transactivator of transcription (Tat) disrupts DA homeostasis, leading to HIV-associated neurocognitive disorders. Analogs of Southern Research Institute 32743, a novel allosteric modulator of the Tat-DA transporter (DAT) interaction, were evaluated in the current study and characterized as atypical ligands of DA uptake. Four novel lead compounds demonstrated high potency to attenuate Tat-induced inhibition of human DAT-mediated DA uptake in an allosteric modulatory manner with no effects on the dynamics of DA uptake-release in DAT.
Collapse
Affiliation(s)
- Ana Catya Jimenez-Torres
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, South Carolina
| | - Jamison A Hastie
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, South Carolina
| | - Sarah E Davis
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, South Carolina
| | - Katherine D Porter
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, South Carolina
| | - Bin Lei
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, South Carolina
| | - Omar Moukha-Chafiq
- Department of Chemistry, Scientific Platforms Division, Southern Research, Birmingham, Alabama
| | - Sixue Zhang
- Department of Chemistry, Scientific Platforms Division, Southern Research, Birmingham, Alabama
| | - Theresa H Nguyen
- Department of Chemistry, Scientific Platforms Division, Southern Research, Birmingham, Alabama
| | - Subramaniam Ananthan
- Department of Chemistry, Scientific Platforms Division, Southern Research, Birmingham, Alabama
| | | | - Jun Zhu
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, South Carolina.
| |
Collapse
|
4
|
Pla-Tenorio J, Velazquez-Perez B, Mendez-Borrero Y, Cruz ML, Sepulveda-Orengo MT, Noel RJ. Astrocytic HIV-1 Nef Expression Decreases Glutamate Transporter Expression in the Nucleus Accumbens and Increases Cocaine-Seeking Behavior in Rats. Pharmaceuticals (Basel) 2025; 18:40. [PMID: 39861103 PMCID: PMC11769493 DOI: 10.3390/ph18010040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/23/2024] [Accepted: 12/27/2024] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Cocaine use disorder is an intersecting issue in populations with HIV-1, further exacerbating the clinical course of the disease and contributing to neurotoxicity and neuroinflammation. Cocaine and HIV neurotoxins play roles in neuronal damage during neuroHIV progression by disrupting glutamate homeostasis in the brain. Even with combined antiretroviral therapy (cART), HIV-1 Nef, an early viral protein expressed in approximately 1% of infected astrocytes, remains a key neurotoxin. This study investigates the relationship among Nef, glutamate homeostasis, and cocaine in the nucleus accumbens (NAc), a critical brain region associated with drug motivation and reward. METHODS Male and female Sprague Dawley rats were used to compare the effects of astrocytic Nef and cocaine by molecular analysis of glutamate transporters, GLT-1 and the cysteine glutamate exchanger (xCT), in the NAc. Behavioral assessments for cocaine self-administration were used to evaluate cocaine-seeking behavior. RESULTS The findings indicate that both cocaine and Nef independently decrease the expression of the glutamate transporter GLT-1 in the NAc. Additionally, rats with astrocytic Nef expression exhibited increased cocaine-seeking behavior but demonstrated sex-dependent molecular differences after the behavioral paradigm. CONCLUSIONS The results suggest that the expression of Nef intensifies cocaine-induced alterations in glutamate homeostasis in the NAc, potentially underlying increased cocaine-seeking behavior. Understanding these interactions better may inform therapeutic strategies for managing cocaine use disorder in HIV-infected individuals.
Collapse
Affiliation(s)
- Jessalyn Pla-Tenorio
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, USA;
| | - Bethzaly Velazquez-Perez
- Department of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Ponce, PR 00716, USA; (B.V.-P.); (Y.M.-B.); (M.L.C.); (M.T.S.-O.)
- Department of Biomedical Sciences, Pontifical Catholic University of Puerto Rico, Ponce, PR 00717, USA
| | - Yainira Mendez-Borrero
- Department of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Ponce, PR 00716, USA; (B.V.-P.); (Y.M.-B.); (M.L.C.); (M.T.S.-O.)
| | - Myrella L. Cruz
- Department of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Ponce, PR 00716, USA; (B.V.-P.); (Y.M.-B.); (M.L.C.); (M.T.S.-O.)
| | - Marian T. Sepulveda-Orengo
- Department of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Ponce, PR 00716, USA; (B.V.-P.); (Y.M.-B.); (M.L.C.); (M.T.S.-O.)
| | - Richard J. Noel
- Department of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Ponce, PR 00716, USA; (B.V.-P.); (Y.M.-B.); (M.L.C.); (M.T.S.-O.)
| |
Collapse
|
5
|
Zhu J, Cirincione AB, Strauss MJ, Davis SE, Eans SO, Tribbitt DK, Alshakhshir N, McLaughlin JP. Impact of HIV-1 tat protein on methamphetamine-induced inhibition of vesicular monoamine transporter2-mediated dopamine transport and methamphetamine conditioned place preference in HIV-1 tat transgenic mice. Eur J Pharmacol 2024; 984:177030. [PMID: 39366503 PMCID: PMC11563864 DOI: 10.1016/j.ejphar.2024.177030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 10/01/2024] [Accepted: 10/02/2024] [Indexed: 10/06/2024]
Abstract
Perturbation of dopamine transmission has been implicated as a contributing factor in HIV-1 associated neurocognitive disorders with concurrent methamphetamine (METH) abuse. We have demonstrated that the HIV-1 protein, transactivator of transcription (Tat), decreases dopamine transport through inhibition of vesicular monoamine transporter2 (VMAT2). This study determined the effects of Tat protein on METH-inhibited VMAT2 function and METH-conditioned place preference (CPP). In vitro exposure of isolated mouse whole brain vesicles to recombinant Tat1-86 or METH displayed a concentration-dependent inhibition of the vesicular [3H]Dopamine uptake, in which a combination of Tat and METH induced a greater reduction of dopamine uptake compared to Tat or METH alone. In vivo, the maximal velocity (Vmax) of vesicular [3H]Dopamine uptake was decreased in inducible Tat transgenic (iTat-tg) mice harvested after treatment with either 21-day doxycycline (Dox) or 14-day METH (3 mg/kg, i.p., daily), whereas these mice treated with both Dox and METH displayed an additive reduction of the Vmax compared to either Tat or METH alone. Moreover, Dox-induced Tat expression increased METH-CPP in an exposure-dependent manner, with iTat-tg mice demonstrating a 2.3-fold potentiation of METH-CPP compared with Tat null control mice upon administration of Dox for 14 days. Furthermore, a 7-day administration of Dox reinstated extinguished METH-CPP. Collectively, these results suggest a synergistic effect of Tat protein and METH on inhibition of VMAT2-mediated DA transport, potentially contributing to potentiation of METH-CPP in iTat-tg mice.
Collapse
Affiliation(s)
- Jun Zhu
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, 29208, USA.
| | - Abagail B Cirincione
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, 29208, USA
| | - Matthew J Strauss
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, 29208, USA
| | - Sarah E Davis
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, 29208, USA
| | - Shainnel O Eans
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Danielle K Tribbitt
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Nadine Alshakhshir
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Jay P McLaughlin
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| |
Collapse
|
6
|
Pla-Tenorio J, Velazquez-Perez B, Mendez-Borrero Y, Cruz-Rentas M, Sepulveda-Orengo M, Noel RJ. Astrocytic HIV-1 Nef expression decreases glutamate transporter expression in the nucleus accumbens and increases cocaine-seeking behavior in rats. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.10.617598. [PMID: 39416088 PMCID: PMC11483060 DOI: 10.1101/2024.10.10.617598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Cocaine use disorder is an intersecting issue in populations with HIV-1, further exacerbating the clinical course of the disease, contributing to neurotoxicity and neuroinflammation. Cocaine and HIV neurotoxins play roles in neuronal damage during neuroHIV progression by disrupting glutamate homeostasis in the brain. Even with cART, HIV-1 Nef, an early viral protein expressed in approximately 1% of infected astrocytes, remains a key neurotoxin. This study investigates the relationship that exists between Nef, glutamate homeostasis, and cocaine in the NAc, a critical brain region associated with drug motivation and reward. Using a rat model, we compared the effects of astrocytic Nef and cocaine by molecular analysis of glutamate transporters in the NAc. We further conducted behavioral assessments for cocaine self-administration to evaluate cocaine-seeking behavior. Our findings indicate that both cocaine and Nef independently decrease the expression of the glutamate transporter GLT-1 in the NAc. Additionally, rats with astrocytic Nef expression exhibited increased cocaine-seeking behavior but demonstrated sex dependent molecular differences after behavioral paradigm. In conclusion, our results suggest the expression of Nef intensifies cocaine-induced alterations in glutamate homeostasis in the NAc, potentially underlying increased cocaine-seeking. Understanding these interactions better may inform therapeutic strategies for managing cocaine use disorder in HIV-infected individuals.
Collapse
Affiliation(s)
- Jessalyn Pla-Tenorio
- Department of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Ponce, Puerto Rico
| | - Bethzaly Velazquez-Perez
- Department of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Ponce, Puerto Rico
- Department of Biomedical Sciences, Pontifical Catholic University of Puerto Rico, Ponce, Puerto Rico
| | - Yainira Mendez-Borrero
- Department of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Ponce, Puerto Rico
| | - Myrella Cruz-Rentas
- Department of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Ponce, Puerto Rico
| | - Marian Sepulveda-Orengo
- Department of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Ponce, Puerto Rico
| | - Richard J. Noel
- Department of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Ponce, Puerto Rico
| |
Collapse
|
7
|
Xie Q, Dasari R, Namba MD, Buck LA, Side CM, Park K, Jackson JG, Barker JM. Astrocytic Regulation of Cocaine Locomotor Sensitization in EcoHIV Infected Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.04.611213. [PMID: 39282274 PMCID: PMC11398419 DOI: 10.1101/2024.09.04.611213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Cocaine use disorder (CUD) is highly comorbid with HIV infection and worsens HIV outcomes. Preclinical research on the outcomes of HIV infection may yield crucial information on neurobehavioral changes resulting from chronic drug exposure in people living with HIV (PLWH). Repeated exposure to cocaine alters behavioral responses to cocaine. This includes development of cocaine locomotor sensitization - or increased locomotor responses to the same doses of cocaine - which depends on nucleus accumbens (NAc) neural plasticity. NAc astrocytes are key regulators of neural activity and plasticity, and their function can be impaired by cocaine exposure and HIV infection, thus implicating them as potential regulators of HIV-induced changes in behavioral response to cocaine. To characterize the effects of HIV infection on cocaine locomotor sensitization, we employed the EcoHIV mouse model to assess changes in locomotor responses after repeated cocaine (10mg/kg) exposure and challenge. EcoHIV infection potentiated expression of cocaine sensitization. We also identified EcoHIV-induced increases in expression of the astrocytic nuclear marker Sox9 selectively in the NAc core. To investigate whether modulation of NAc astrocytes could reverse EcoHIV-induced deficits, we employed a chemogenetic approach. We found that chemogenetic activation of NAc astrocyte Gq signaling attenuated EcoHIV-enhanced cocaine sensitization. We propose that HIV infection contributes to cocaine behavioral sensitization and induces adaptations in NAc astrocytes, while promoting NAc astrocytic Gq-signaling can recover EcoHIV-induced behavioral changes. These findings identify potential cellular substrates of disordered cocaine-driven behavior in the context of HIV infection and point toward strategies to reduce cocaine-related behavior in PLWH.
Collapse
|
8
|
Namba MD, Xie Q, Park K, Jackson JG, Barker JM. EcoHIV Infection Modulates the Effects of Cocaine Exposure Pattern and Abstinence on Cocaine Seeking and Neuroimmune Protein Expression in Male Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.15.589615. [PMID: 38659915 PMCID: PMC11042347 DOI: 10.1101/2024.04.15.589615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Cocaine use disorders (CUDs) and human immunodeficiency virus (HIV) remain persistent public health dilemmas throughout the world. One major hurdle for treating CUD is the increase in cocaine craving and seeking behavior that occurs over a protracted period of abstinence, an effect known as the incubation of craving. Little is known about how HIV may modulate this process. Thus, we sought to examine the impact of chronic HIV infection on the incubation of cocaine craving and associated changes in the central and peripheral immune systems. Here, mice were inoculated with EcoHIV, which is a chimeric HIV-1 construct that produces chronic HIV infection in mice. EcoHIV- and sham-infected mice were conditioned with cocaine daily or intermittently in a conditioned place preference (CPP) paradigm, followed by 1 or 21 days of forced abstinence prior to assessing preference for the cocaine-paired chamber. Under both conditioning regimens, sham mice exhibited incubation of cocaine CPP after 21 days of abstinence. EcoHIV-infected mice conditioned daily with cocaine showed enhanced cocaine seeking at both abstinence timepoints, whereas infected mice conditioned intermittently showed a reversal of the incubation effect, with higher cocaine seeking after 1 day of abstinence compared to 21 days. Analysis of corticolimbic CX3CL1-CX3CR1 and glutamate receptor expression revealed alterations in medial prefrontal cortex (mPFC) CX3CL1 and nucleus accumbens (NAc) GluN2A receptors that correlated with cocaine seeking following daily cocaine exposure. Moreover, examination of peripheral immune markers showed that the effect of abstinence and EcoHIV infection on these measures depended on the cocaine exposure regimen. Altogether, these results highlight the importance of cocaine abstinence and exposure pattern as critical variables that modulate HIV-associated neuroimmune outcomes and relapse vulnerability.
Collapse
Affiliation(s)
- Mark D. Namba
- Department of Pharmacology & Physiology, College of Medicine, Drexel University, Philadelphia, PA, USA
| | - Qiaowei Xie
- Department of Pharmacology & Physiology, College of Medicine, Drexel University, Philadelphia, PA, USA
- Graduate Program in Pharmacology and Physiology, College of Medicine, Drexel University
| | - Kyewon Park
- Center for AIDS Research (CFAR), University of Pennsylvania, Philadelphia, PA, USA
| | - Joshua G. Jackson
- Department of Pharmacology & Physiology, College of Medicine, Drexel University, Philadelphia, PA, USA
| | - Jacqueline M. Barker
- Department of Pharmacology & Physiology, College of Medicine, Drexel University, Philadelphia, PA, USA
| |
Collapse
|
9
|
Buck LA, Xie Q, Willis M, Side CM, Giacometti LL, Gaskill PJ, Park K, Shaheen F, Guo L, Gorantla S, Barker JM. Impaired extinction of cocaine seeking in HIV-infected mice is accompanied by peripheral and central immune dysregulation. Commun Biol 2024; 7:387. [PMID: 38553542 PMCID: PMC10980811 DOI: 10.1038/s42003-024-06079-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 03/20/2024] [Indexed: 04/01/2024] Open
Abstract
Substance use disorders (SUDs) are highly comorbid with HIV infection, necessitating an understanding of the interactive effects of drug exposure and HIV. The relationship between HIV infection and cocaine use disorder is likely bidirectional, with cocaine use directly impacting immune function while HIV infection alters addiction-related behavior. To better characterize the neurobehavioral and immune consequences of HIV infection and cocaine exposure, this study utilizes a humanized mouse model to investigate the outcomes of HIV-1 infection on cocaine-related behaviors in a conditioned place preference (CPP) model, and the interactive effects of cocaine and HIV infection on peripheral and central nervous system inflammation. HIV infection selectively impairs cocaine CPP extinction without effecting reinstatement or cocaine seeking under conflict. Behavioral alterations are accompanied by immune changes in HIV infected mice, including increased prefrontal cortex astrocyte immunoreactivity and brain-region specific effects on microglia number and reactivity. Peripheral immune system changes are observed in human cytokines, including HIV-induced reductions in human TNFα, and cocaine and HIV interactions on GM-CSF levels. Together these data provide new insights into the unique neurobehavioral outcomes of HIV infection and cocaine exposure and how they interact to effect immune responses.
Collapse
Affiliation(s)
- Lauren A Buck
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Qiaowei Xie
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, USA
- Graduate Program in Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Michelle Willis
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Christine M Side
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Laura L Giacometti
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Peter J Gaskill
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Kyewon Park
- Center for AIDS Research, University of Pennsylvania, Philadelphia, PA, USA
| | - Farida Shaheen
- Center for AIDS Research, University of Pennsylvania, Philadelphia, PA, USA
| | - Lili Guo
- Medical Center, University of Nebraska, Omaha, NE, USA
| | | | - Jacqueline M Barker
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
10
|
Vines L, Sotelo D, Giddens N, Manza P, Volkow ND, Wang GJ. Neurological, Behavioral, and Pathophysiological Characterization of the Co-Occurrence of Substance Use and HIV: A Narrative Review. Brain Sci 2023; 13:1480. [PMID: 37891847 PMCID: PMC10605099 DOI: 10.3390/brainsci13101480] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/10/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
Combined antiretroviral therapy (cART) has greatly reduced the severity of HIV-associated neurocognitive disorders in people living with HIV (PLWH); however, PLWH are more likely than the general population to use drugs and suffer from substance use disorders (SUDs) and to exhibit risky behaviors that promote HIV transmission and other infections. Dopamine-boosting psychostimulants such as cocaine and methamphetamine are some of the most widely used substances among PLWH. Chronic use of these substances disrupts brain function, structure, and cognition. PLWH with SUD have poor health outcomes driven by complex interactions between biological, neurocognitive, and social factors. Here we review the effects of comorbid HIV and psychostimulant use disorders by discussing the distinct and common effects of HIV and chronic cocaine and methamphetamine use on behavioral and neurological impairments using evidence from rodent models of HIV-associated neurocognitive impairments (Tat or gp120 protein expression) and clinical studies. We also provide a biopsychosocial perspective by discussing behavioral impairment in differentially impacted social groups and proposing interventions at both patient and population levels.
Collapse
Affiliation(s)
- Leah Vines
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA; (L.V.); (D.S.); (P.M.); (N.D.V.)
| | - Diana Sotelo
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA; (L.V.); (D.S.); (P.M.); (N.D.V.)
| | - Natasha Giddens
- Department of Psychiatry, University of Wisconsin School of Medicine and Public Health, Madison, WI 53719, USA;
| | - Peter Manza
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA; (L.V.); (D.S.); (P.M.); (N.D.V.)
| | - Nora D. Volkow
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA; (L.V.); (D.S.); (P.M.); (N.D.V.)
| | - Gene-Jack Wang
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA; (L.V.); (D.S.); (P.M.); (N.D.V.)
| |
Collapse
|
11
|
Namba MD, Xie Q, Barker JM. Advancing the preclinical study of comorbid neuroHIV and substance use disorders: Current perspectives and future directions. Brain Behav Immun 2023; 113:453-475. [PMID: 37567486 PMCID: PMC10528352 DOI: 10.1016/j.bbi.2023.07.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 06/23/2023] [Accepted: 07/30/2023] [Indexed: 08/13/2023] Open
Abstract
Human immunodeficiency virus (HIV) remains a persistent public health concern throughout the world. Substance use disorders (SUDs) are a common comorbidity that can worsen treatment outcomes for people living with HIV. The relationship between HIV infection and SUD outcomes is likely bidirectional, making clear interrogation of neurobehavioral outcomes challenging in clinical populations. Importantly, the mechanisms through which HIV and addictive drugs disrupt homeostatic immune and CNS function appear to be highly overlapping and synergistic within HIV-susceptible reward and motivation circuitry in the central nervous system. Decades of animal research have revealed invaluable insights into mechanisms underlying the pathophysiology SUDs and HIV, although translational studies examining comorbid SUDs and HIV are very limited due to the technical challenges of modeling HIV infection preclinically. In this review, we discuss preclinical animal models of HIV and highlight key pathophysiological characteristics of each model, with a particular emphasis on rodent models of HIV. We then review the implementation of these models in preclinical SUD research and identify key gaps in knowledge in the field. Finally, we discuss how cutting-edge behavioral neuroscience tools, which have revealed key insights into the neurobehavioral mechanisms of SUDs, can be applied to preclinical animal models of HIV to reveal potential, novel treatment avenues for comorbid HIV and SUDs. Here, we argue that future preclinical SUD research would benefit from incorporating comorbidities such as HIV into animal models and would facilitate the discovery of more refined, subpopulation-specific mechanisms and effective SUD prevention and treatment targets.
Collapse
Affiliation(s)
- Mark D Namba
- Department of Pharmacology & Physiology, College of Medicine, Drexel University, Philadelphia, PA, USA
| | - Qiaowei Xie
- Department of Pharmacology & Physiology, College of Medicine, Drexel University, Philadelphia, PA, USA
| | - Jacqueline M Barker
- Department of Pharmacology & Physiology, College of Medicine, Drexel University, Philadelphia, PA, USA.
| |
Collapse
|
12
|
Barker J, Buck L, Xie Q, Willis M, Side C, Giacometti L, Gaskill P, Park K, Shaheen F, Guo L, Gorantla S. Impaired extinction of cocaine seeking in HIV-infected mice is accompanied by peripheral and central immune dysregulation. RESEARCH SQUARE 2023:rs.3.rs-3276379. [PMID: 37841842 PMCID: PMC10571607 DOI: 10.21203/rs.3.rs-3276379/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
Substance use disorders (SUDs) are highly comorbid with HIV infection, necessitating an understanding of the interactive effects of drug exposure and HIV. The relationship between HIV infection and cocaine use disorder is likely bidirectional, with cocaine use directly impacting immune function while HIV infection alters addiction-related behavior. To better characterize the neurobehavioral and immune consequences of HIV infection and cocaine exposure, this study utilized a humanized mouse model to investigate the outcomes of HIV-1 infection on cocaine-related behaviors in a conditioned place preference (CPP) model, and the interactive effects of cocaine and HIV infection on peripheral and central nervous system inflammation. HIV infection selectively impaired cocaine CPP extinction without effecting reinstatement or cocaine seeking under conflict were observed. Behavioral alterations were accompanied by immune changes in HIV infected mice, including increased prefrontal cortex astrocyte immunoreactivity and brain-region specific effects on microglia number and reactivity. Peripheral immune system changes were observed in both mouse and human cytokines, including HIV-induced reductions in mouse IL-1α and G-CSF and human TNFα, and cocaine induced alterations in mouse GM-CSF. Together these data provide new insights into the unique neurobehavioral outcomes of HIV infection and cocaine exposure and how they interact to effect immune responses.
Collapse
|
13
|
Pla-Tenorio J, Roig AM, García-Cesaní PA, Santiago LA, Sepulveda-Orengo MT, Noel RJ. Astrocytes: Role in pathogenesis and effect of commonly misused drugs in the HIV infected brain. CURRENT RESEARCH IN NEUROBIOLOGY 2023; 5:100108. [PMID: 38020814 PMCID: PMC10663134 DOI: 10.1016/j.crneur.2023.100108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 06/05/2023] [Accepted: 08/18/2023] [Indexed: 12/01/2023] Open
Abstract
The roles of astrocytes as reservoirs and producers of a subset of viral proteins in the HIV infected brain have been studied extensively as a key to understanding HIV-associated neurocognitive disorders (HAND). However, their comprehensive role in the context of intersecting substance use and neurocircuitry of the reward pathway and HAND has yet to be fully explained. Use of methamphetamines, cocaine, or opioids in the context of HIV infection have been shown to lead to a faster progression of HAND. Glutamatergic, dopaminergic, and GABAergic systems are implicated in the development of HAND-induced cognitive impairments. A thorough review of scientific literature exploring the variety of mechanisms in which these drugs exert their effects on the HIV brain and astrocytes has revealed marked areas of convergence in overexcitation leading to increased drug-seeking behavior, inflammation, apoptosis, and irreversible neurotoxicity. The present review investigates astrocytes, the neural pathways, and mechanisms of drug disruption that ultimately play a larger holistic role in terms of HIV progression and drug use. There are opportunities for future research, therapeutic intervention, and preventive strategies to diminish HAND in the subset population of patients with HIV and substance use disorder.
Collapse
Affiliation(s)
- Jessalyn Pla-Tenorio
- Ponce Health Sciences University, School of Medicine, Department of Basic Sciences, 395 Industrial Reparada, Zona 2, Ponce, PR, 00716, Puerto Rico
| | - Angela M. Roig
- Seattle Children's Hospital, MS OC.7.830, 4800 Sand Point Way NE, Seattle, WA, 98105-0371, United States
| | - Paulina A. García-Cesaní
- Bella Vista Hospital, Family Medicine Residency, Carr. 349 Km 2.7, Cerro Las Mesas, Mayaguez, PR, 00681, Puerto Rico
| | - Luis A. Santiago
- Ponce Health Sciences University, School of Medicine, Department of Basic Sciences, 395 Industrial Reparada, Zona 2, Ponce, PR, 00716, Puerto Rico
| | - Marian T. Sepulveda-Orengo
- Ponce Health Sciences University, School of Medicine, Department of Basic Sciences, 395 Industrial Reparada, Zona 2, Ponce, PR, 00716, Puerto Rico
| | - Richard J. Noel
- Ponce Health Sciences University, School of Medicine, Department of Basic Sciences, 395 Industrial Reparada, Zona 2, Ponce, PR, 00716, Puerto Rico
| |
Collapse
|
14
|
Buck LA, Xie Q, Willis M, Side CM, Giacometti LL, Gaskill PJ, Park K, Shaheen F, Guo L, Gorantla S, Barker JM. Impaired extinction of cocaine seeking in HIV-infected mice is accompanied by peripheral and central immune dysregulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.11.552858. [PMID: 37645889 PMCID: PMC10462035 DOI: 10.1101/2023.08.11.552858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Substance use disorders (SUDs) are highly comorbid with HIV infection, necessitating an understanding of the interactive effects of drug exposure and HIV. The relationship between progressive HIV infection and cocaine use disorder is likely bidirectional, with cocaine use having direct effects on immune function while HIV infection can alter addiction-related behavior. To better characterized the neurobehavioral and immune consequences of HIV infection and cocaine exposure, this study utilized a humanized mouse model to investigate the outcomes of progressive HIV infection on cocaine-related behaviors in a cocaine conditioned place preference (CPP) model, and the interactive effects of cocaine and HIV infection on peripheral and central nervous system inflammation. HIV infection did not impact the formation of a cocaine CPP, but did result in resistance to extinction of the CPP. No effects of HIV on yohimbine-primed reinstatement or cocaine seeking under conflict were observed. These behavioral alterations were accompanied by immune changes in HIV infected mice, including increased prefrontal cortex astrocyte immunoreactivity and brain-region specific effects on microglia number and reactivity. Peripheral immune system changes were observed in both mouse and human markers. Among other targets, this included HIV-induced reductions in mouse IL-1α and G-CSF and human TNFα and cocaine-induced alterations in human TNFα and mouse GM-CSF such that cocaine exposure increases both cytokines only in the absence of HIV infection. Together these data provide new insights into the unique neurobehavioral processes underlying HIV infection and cocaine use disorders, and further how they interact to effect immune responses.
Collapse
|
15
|
Namba MD, Phillips MN, Chen PJ, Blass BE, Olive MF, Neisewander JL. HIV gp120 impairs nucleus accumbens neuroimmune function and dopamine D3 receptor-mediated inhibition of cocaine seeking in male rats. ADDICTION NEUROSCIENCE 2023; 5:100062. [PMID: 36909738 PMCID: PMC9997483 DOI: 10.1016/j.addicn.2023.100062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Cocaine Use Disorders (CUDs) are associated with an increased risk of human immunodeficiency virus (HIV) infection. Cocaine and the HIV envelope protein gp120 each induce distinct deficits to mesocorticolimbic circuit function and motivated behavior; however, little is known regarding how they interact to dysregulate these functions or how such interactions impact pharmacotherapeutic efficacy. We have previously shown that the selective, weak partial agonist of the dopamine D3 receptor (D3R), MC-25-41, attenuates cocaine-seeking behavior in male rats. Here, we sought to characterize changes in striatal neuroimmune function in gp120-exposed rats across abstinence from operant access to cocaine (0.75 mg/kg, i.v.) or sucrose (45 mg/pellet), and to examine the impact of gp120 exposure on MC-25-41-reduced cocaine seeking. After establishing a history of cocaine or sucrose self-administration, rats received intracerebroventricular gp120 infusions daily the first 5 days of abstinence and were sacrificed either on day 6 or after 21 days of forced abstinence and a cue-induced cocaine seeking test. We demonstrated that MC-25-41 treatment attenuated cue-induced cocaine seeking among control rats but not gp120-exposed rats. Moreover, postmortem analysis of nucleus accumbens (NAc) core neuroimmune function indicated cocaine abstinence- and gp120-induced impairments, and the expression of several immune factors within the NAc core significantly correlated with cocaine-seeking behavior. We conclude that cocaine abstinence dysregulates striatal neuroimmune function and interacts with gp120 to inhibit the effectiveness of a D3R partial agonist in reducing cocaine seeking. These findings highlight the need to consider comorbidities, such as immune status, when evaluating the efficacy of novel pharmacotherapeutics.
Collapse
Affiliation(s)
- Mark D Namba
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Megan N Phillips
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Peng-Jen Chen
- Department of Pharmaceutical Sciences, Moulder Center for Drug Discovery Research, School of Pharmacy, Temple University, Philadelphia, PA, USA
| | - Benjamin E Blass
- Department of Pharmaceutical Sciences, Moulder Center for Drug Discovery Research, School of Pharmacy, Temple University, Philadelphia, PA, USA
| | - M Foster Olive
- Department of Psychology, Arizona State University, Tempe, AZ, USA
| | | |
Collapse
|
16
|
Akins NS, Salahuddin MF, Pandey P, Kim SJ, Mahdi F, Khan MIH, Moss EM, Worth CJ, Keane MM, Chittiboyina AG, Doerksen RJ, Paris JJ, Le HV. Alleviation of Cocaine Withdrawal and Pertinent Interactions between Salvinorin-Based Antagonists and Kappa Opioid Receptor. ACS Chem Neurosci 2023; 14:958-976. [PMID: 36795782 DOI: 10.1021/acschemneuro.2c00806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023] Open
Abstract
The kappa opioid receptor (KOR) is involved in the regulation of both the reward and mood processes. Recent reports find that the use of drugs of abuse increases the production of dynorphin and the overall activation of KOR. Long-acting KOR antagonists, such as norbinaltorphimine (nor-BNI), JDTic, and 5'-guanidinonaltrindole (GNTI), have been shown to stop depressive and anxiety-related disorders, which are the common side effects of withdrawal that can lead to a relapse in drug use. Unfortunately, these prototypical KOR antagonists are known to induce selective KOR antagonism that is delayed by hours and extremely prolonged, and their use in humans comes with serious safety concerns because they possess a large window for potential drug-drug interactions. Furthermore, their persistent pharmacodynamic activities can hinder the ability to reverse unanticipated side effects immediately. Herein, we report our studies of the lead selective, salvinorin-based KOR antagonist (1) as well as nor-BNI on C57BL/6N male mice for spontaneous cocaine withdrawal. Assessment of pharmacokinetics showed that 1 is a short-acting compound with an average half-life of 3.75 h across different compartments (brain, spinal cord, liver, and plasma). Both 1 (5 mg/kg) and nor-BNI (5 mg/kg) were shown to reduce spontaneous withdrawal behavior in mice, with 1 producing additional anti-anxiety-like behavior in a light-dark transition test (however, no mood-related effects of 1 or nor-BNI were observed at the current dosing in an elevated plus maze or a tail suspension test). Our results support the study of selective, short-acting KOR antagonists for the treatment of psychostimulant withdrawal and the associated negative mood states that contribute to relapse. Furthermore, we identified pertinent interactions between 1 and KOR via computational studies, including induced-fit docking, mutagenesis, and molecular dynamics simulations, to gain insight into the design of future selective, potent, and short-acting salvinorin-based KOR antagonists.
Collapse
Affiliation(s)
- Nicholas S Akins
- Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, University, Mississippi 38677, United States
| | - Mohammed F Salahuddin
- Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, University, Mississippi 38677, United States
| | - Pankaj Pandey
- National Center for Natural Products Research, School of Pharmacy, University of Mississippi, University, Mississippi 38677, United States
| | - Seong Jong Kim
- Natural Products Utilization Research Unit, United States Department of Agriculture, Agricultural Research Service, University, Mississippi 38677, United States
| | - Fakhri Mahdi
- Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, University, Mississippi 38677, United States
| | - Md Imdadul H Khan
- Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, University, Mississippi 38677, United States
| | - Emaya M Moss
- Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, University, Mississippi 38677, United States
| | - Charlie J Worth
- Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, University, Mississippi 38677, United States
| | - Madeline M Keane
- Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, University, Mississippi 38677, United States
| | - Amar G Chittiboyina
- National Center for Natural Products Research, School of Pharmacy, University of Mississippi, University, Mississippi 38677, United States
| | - Robert J Doerksen
- Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, University, Mississippi 38677, United States.,Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, Mississippi 38677, United States
| | - Jason J Paris
- Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, University, Mississippi 38677, United States.,Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, Mississippi 38677, United States
| | - Hoang V Le
- Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, University, Mississippi 38677, United States.,Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, Mississippi 38677, United States
| |
Collapse
|
17
|
Zhu J, Quizon PM, Wang Y, Adeniran CA, Strauss MJ, Jiménez-Torres AC, Patel P, Cirino TJ, Eans SO, Hammond HR, Deliscar LS, O'Hara P, Saini SK, Ofori E, Vekariya RH, Zhang S, Moukha-Chafiq O, Nguyen TH, Ananthan S, Augelli-Szafran CE, Zhan CG, McLaughlin JP. SRI-32743, a novel allosteric modulator, attenuates HIV-1 Tat protein-induced inhibition of the dopamine transporter and alleviates the potentiation of cocaine reward in HIV-1 Tat transgenic mice. Neuropharmacology 2022; 220:109239. [PMID: 36126727 DOI: 10.1016/j.neuropharm.2022.109239] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 08/09/2022] [Accepted: 09/01/2022] [Indexed: 11/21/2022]
Abstract
Cocaine abuse increases the incidence of HIV-1-associated neurocognitive disorders. We have demonstrated that HIV-1 transactivator of transcription (Tat) allosterically modulates dopamine (DA) reuptake through the human DA transporter (hDAT), potentially contributing to Tat-induced cognitive impairment and potentiation of cocaine conditioned place preference (CPP). This study determined the effects of a novel allosteric modulator of DAT, SRI-32743, on the interactions of HIV-1 Tat, DA, cocaine, and [3H]WIN35,428 with hDAT in vitro. SRI-32743 (50 nM) attenuated Tat-induced inhibition of [3H]DA uptake and decreased the cocaine-mediated dissociation of [3H]WIN35,428 binding in CHO cells expressing hDAT, suggesting a SRI-32743-mediated allosteric modulation of the Tat-DAT interaction. In further in vivo studies utilizing doxycycline-inducible Tat transgenic (iTat-tg) mice, 14 days of Tat expression significantly reduced the recognition index by 31.7% in the final phase of novel object recognition (NOR) and potentiated cocaine-CPP 2.7-fold compared to responses of vehicle-treated control iTat-tg mice. The Tat-induced NOR deficits and potentiation of cocaine-CPP were not observed in saline-treated iTat-tg or doxycycline-treated G-tg (Tat-null) mice. Systemic administration (i.p.) of SRI-32743 prior to behavioral testing ameliorated Tat-induced impairment of NOR (at a dose of 10 mg/kg) and the Tat-induced potentiation of cocaine-CPP (at doses of 1 or 10 mg/kg). These findings demonstrate that Tat and cocaine interactions with DAT may be regulated by compounds interacting at the DAT allosteric modulatory sites, suggesting a potential therapeutic intervention for HIV-infected patients with concurrent cocaine abuse.
Collapse
Affiliation(s)
- Jun Zhu
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA.
| | - Pamela M Quizon
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Yingying Wang
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Charles A Adeniran
- Molecular Modeling and Biopharmaceutical Center, USA; Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA
| | - Matthew J Strauss
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Ana C Jiménez-Torres
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Palak Patel
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Thomas J Cirino
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32611, USA
| | - Shainnel O Eans
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32611, USA
| | - Haylee R Hammond
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32611, USA
| | - Laure S Deliscar
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32611, USA
| | - Priscilla O'Hara
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32611, USA
| | - Surendra K Saini
- Department of Chemistry, Scientific Platforms, Southern Research, Birmingham, AL 35205, USA
| | - Edward Ofori
- Department of Chemistry, Scientific Platforms, Southern Research, Birmingham, AL 35205, USA
| | - Rakesh H Vekariya
- Department of Chemistry, Scientific Platforms, Southern Research, Birmingham, AL 35205, USA
| | - Sixue Zhang
- Department of Chemistry, Scientific Platforms, Southern Research, Birmingham, AL 35205, USA
| | - Omar Moukha-Chafiq
- Department of Chemistry, Scientific Platforms, Southern Research, Birmingham, AL 35205, USA
| | - Theresa H Nguyen
- Department of Chemistry, Scientific Platforms, Southern Research, Birmingham, AL 35205, USA
| | - Subramaniam Ananthan
- Department of Chemistry, Scientific Platforms, Southern Research, Birmingham, AL 35205, USA
| | | | - Chang-Guo Zhan
- Molecular Modeling and Biopharmaceutical Center, USA; Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA
| | - Jay P McLaughlin
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
18
|
Zhao X, Zhang F, Kandel SR, Brau F, He JJ. HIV Tat and cocaine interactively alter genome-wide DNA methylation and gene expression and exacerbate learning and memory impairments. Cell Rep 2022; 39:110765. [PMID: 35508123 PMCID: PMC9615417 DOI: 10.1016/j.celrep.2022.110765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 03/16/2022] [Accepted: 04/07/2022] [Indexed: 11/03/2022] Open
Abstract
Cocaine use is a major comorbidity of HIV-associated neurocognitive disorder (HAND). In this study, we show that cocaine exposure worsens the learning and memory of doxycycline-inducible and brain-specific HIV Tat transgenic mice (iTat) and results in 14,838 hypermethylated CpG-related differentially methylated regions (DMRs) and 15,800 hypomethylated CpG-related DMRs, which are linked to 52 down- and 127 upregulated genes, respectively, in the hippocampus of iTat mice. These genes are mostly enriched at the neuronal function-, cell morphology-, and synapse formation-related extracellular matrix (ECM) receptor-ligand interaction pathway and mostly impacted in microglia. The accompanying neuropathological changes include swollen dendritic spines, increased synaptophysin expression, and diminished glial activation. We also find that sex (female) and age additively worsen the behavioral and pathological changes. These findings together indicate that chronic cocaine and long-term Tat expression interactively contribute to HAND, likely involving changes of DNA methylation and ECM receptor-ligand interactions.
Collapse
Affiliation(s)
- Xiaojie Zhao
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University, North Chicago, IL 60064, USA; Center for Cancer Cell Biology, Immunology and Infection, Rosalind Franklin University, North Chicago, IL 60064, USA; School of Graduate and Postdoctoral Studies, Rosalind Franklin University, North Chicago, IL 60064, USA
| | - Fan Zhang
- Department of Family Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Suresh R Kandel
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University, North Chicago, IL 60064, USA; Center for Cancer Cell Biology, Immunology and Infection, Rosalind Franklin University, North Chicago, IL 60064, USA
| | - Frédéric Brau
- Université Côte d'Azur, CNRS, IPMC, Sophia-Antipolis 06560, France
| | - Johnny J He
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University, North Chicago, IL 60064, USA; Center for Cancer Cell Biology, Immunology and Infection, Rosalind Franklin University, North Chicago, IL 60064, USA; School of Graduate and Postdoctoral Studies, Rosalind Franklin University, North Chicago, IL 60064, USA.
| |
Collapse
|
19
|
Davis S, Zhu J. Substance abuse and neurotransmission. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2022; 93:403-441. [PMID: 35341573 PMCID: PMC9759822 DOI: 10.1016/bs.apha.2021.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The number of people who suffer from a substance abuse disorder has continued to rise over the last decade; particularly, the number of drug-related overdose deaths has sharply increased during the COVID-19 pandemic. Converging lines of clinical observations, supported by imaging and neuropsychological performance testing, have demonstrated that substance abuse-induced dysregulation of neurotransmissions in the brain is critical for development and expression of the addictive properties of abused substances. Recent scientific advances have allowed for better understanding of the neurobiological processes that mediates drugs of abuse and addiction. This chapter presents the past classic concepts and the recent advances in our knowledge about how cocaine, amphetamines, opioids, alcohol, and nicotine alter multiple neurotransmitter systems, which contribute to the behaviors associated with each drug. Additionally, we discuss the interactive effects of HIV-1 or COVID-19 and substance abuse on neurotransmission and neurobiological pathways. Finally, we introduce therapeutic strategies for development of pharmacotherapies for substance abuse disorders.
Collapse
Affiliation(s)
- Sarah Davis
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, United States
| | - Jun Zhu
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, United States.
| |
Collapse
|
20
|
League AF, Gorman BL, Hermes DJ, Johnson CT, Jacobs IR, Yadav-Samudrala BJ, Poklis JL, Niphakis MJ, Cravatt BF, Lichtman AH, Ignatowska-Jankowska BM, Fitting S. Monoacylglycerol Lipase Inhibitor MJN110 Reduces Neuronal Hyperexcitability, Restores Dendritic Arborization Complexity, and Regulates Reward-Related Behavior in Presence of HIV-1 Tat. Front Neurol 2021; 12:651272. [PMID: 34484091 PMCID: PMC8415271 DOI: 10.3389/fneur.2021.651272] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 07/12/2021] [Indexed: 12/01/2022] Open
Abstract
While current therapeutic strategies for people living with human immunodeficiency virus type 1 (HIV-1) suppress virus replication peripherally, viral proteins such as transactivator of transcription (Tat) enter the central nervous system early upon infection and contribute to chronic inflammatory conditions even alongside antiretroviral treatment. As demand grows for supplemental strategies to combat virus-associated pathology presenting frequently as HIV-associated neurocognitive disorders (HAND), the present study aimed to characterize the potential utility of inhibiting monoacylglycerol lipase (MAGL) activity to increase inhibitory activity at cannabinoid receptor-type 1 receptors through upregulation of 2-arachidonoylglycerol (2-AG) and downregulation of its degradation into proinflammatory metabolite arachidonic acid (AA). The MAGL inhibitor MJN110 significantly reduced intracellular calcium and increased dendritic branching complexity in Tat-treated primary frontal cortex neuron cultures. Chronic MJN110 administration in vivo increased 2-AG levels in the prefrontal cortex (PFC) and striatum across Tat(+) and Tat(–) groups and restored PFC N-arachidonoylethanolamine (AEA) levels in Tat(+) subjects. While Tat expression significantly increased rate of reward-related behavioral task acquisition in a novel discriminative stimulus learning and cognitive flexibility assay, MJN110 altered reversal acquisition specifically in Tat(+) mice to rates indistinguishable from Tat(–) controls. Collectively, our results suggest a neuroprotective role of MAGL inhibition in reducing neuronal hyperexcitability, restoring dendritic arborization complexity, and mitigating neurocognitive alterations driven by viral proteins associated with latent HIV-1 infection.
Collapse
Affiliation(s)
- Alexis F League
- Department of Psychology and Neuroscience, University of North Carolina Chapel Hill, Chapel Hill, NC, United States
| | - Benjamin L Gorman
- Department of Psychology and Neuroscience, University of North Carolina Chapel Hill, Chapel Hill, NC, United States
| | - Douglas J Hermes
- Department of Psychology and Neuroscience, University of North Carolina Chapel Hill, Chapel Hill, NC, United States
| | - Clare T Johnson
- Department of Psychology and Neuroscience, University of North Carolina Chapel Hill, Chapel Hill, NC, United States
| | - Ian R Jacobs
- Department of Psychology and Neuroscience, University of North Carolina Chapel Hill, Chapel Hill, NC, United States
| | - Barkha J Yadav-Samudrala
- Department of Psychology and Neuroscience, University of North Carolina Chapel Hill, Chapel Hill, NC, United States
| | - Justin L Poklis
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States
| | - Micah J Niphakis
- Department of Chemistry, Scripps Research Institute, La Jolla, CA, United States
| | - Benjamin F Cravatt
- Department of Chemistry, Scripps Research Institute, La Jolla, CA, United States
| | - Aron H Lichtman
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States
| | | | - Sylvia Fitting
- Department of Psychology and Neuroscience, University of North Carolina Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
21
|
Namba MD, Leyrer-Jackson JM, Nagy EK, Olive MF, Neisewander JL. Neuroimmune Mechanisms as Novel Treatment Targets for Substance Use Disorders and Associated Comorbidities. Front Neurosci 2021; 15:650785. [PMID: 33935636 PMCID: PMC8082184 DOI: 10.3389/fnins.2021.650785] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/24/2021] [Indexed: 12/12/2022] Open
Abstract
Recent studies examining the neurobiology of substance abuse have revealed a significant role of neuroimmune signaling as a mechanism through which drugs of abuse induce aberrant changes in synaptic plasticity and contribute to substance abuse-related behaviors. Immune signaling within the brain and the periphery critically regulates homeostasis of the nervous system. Perturbations in immune signaling can induce neuroinflammation or immunosuppression, which dysregulate nervous system function including neural processes associated with substance use disorders (SUDs). In this review, we discuss the literature that demonstrates a role of neuroimmune signaling in regulating learning, memory, and synaptic plasticity, emphasizing specific cytokine signaling within the central nervous system. We then highlight recent preclinical studies, within the last 5 years when possible, that have identified immune mechanisms within the brain and the periphery associated with addiction-related behaviors. Findings thus far underscore the need for future investigations into the clinical potential of immunopharmacology as a novel approach toward treating SUDs. Considering the high prevalence rate of comorbidities among those with SUDs, we also discuss neuroimmune mechanisms of common comorbidities associated with SUDs and highlight potentially novel treatment targets for these comorbid conditions. We argue that immunopharmacology represents a novel frontier in the development of new pharmacotherapies that promote long-term abstinence from drug use and minimize the detrimental impact of SUD comorbidities on patient health and treatment outcomes.
Collapse
Affiliation(s)
- Mark D. Namba
- School of Life Sciences, Arizona State University, Tempe, AZ, United States
| | | | - Erin K. Nagy
- Department of Psychology, Arizona State University, Tempe, AZ, United States
| | - M. Foster Olive
- Department of Psychology, Arizona State University, Tempe, AZ, United States
| | | |
Collapse
|
22
|
Cirino TJ, McLaughlin JP. Mini review: Promotion of substance abuse in HIV patients: Biological mediation by HIV-1 Tat protein. Neurosci Lett 2021; 753:135877. [PMID: 33838257 DOI: 10.1016/j.neulet.2021.135877] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 03/28/2021] [Accepted: 03/30/2021] [Indexed: 11/29/2022]
Abstract
Despite successful viral suppression by combinatorial anti-retroviral therapy, HIV infection continues to negatively impact the quality of life of patients by promoting neuropathy and HIV-Associated Neurocognitive Disorders (HAND), where substance use disorder (SUD) is highly comorbid and known to worsen health outcomes. While substance abuse exacerbates the progression of HIV, emerging evidence also suggests the virus may potentiate the rewarding effect of abused substances. As HIV does not infect neurons, these effects are theorized to be mediated by viral proteins. Key among these proteins are HIV-1 Tat, which can continue to be produced under viral suppression in patients. This review will recap the behavioral evidence for HIV-1 Tat mediation of a potentiation of cocaine, opioid and alcohol reward, and explore the neurochemical dysfunction associated by Tat as potential mechanisms underlying changes in reward. Targeting rampant oxidative stress, inflammation and excitotoxicity associated with HIV and Tat protein exposure may prove useful in combating persistent substance abuse comorbid with HIV in the clinic.
Collapse
Affiliation(s)
- Thomas J Cirino
- Department of Neurology, School of Medicine, University of California at San Francisco, San Francisco, CA, 94158, USA
| | - Jay P McLaughlin
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
23
|
Sil S, Thangaraj A, Chivero ET, Niu F, Kannan M, Liao K, Silverstein PS, Periyasamy P, Buch S. HIV-1 and drug abuse comorbidity: Lessons learned from the animal models of NeuroHIV. Neurosci Lett 2021; 754:135863. [PMID: 33794296 DOI: 10.1016/j.neulet.2021.135863] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 03/23/2021] [Accepted: 03/25/2021] [Indexed: 02/06/2023]
Abstract
Various research studies that have investigated the association between HIV infection and addiction underpin the role of various drugs of abuse in impairing immunological and non-immunological pathways of the host system, ultimately leading to augmentation of HIV infection and disease progression. These studies have included both in vitro and in vivo animal models wherein investigators have assessed the effects of various drugs on several disease parameters to decipher the impact of drugs on both HIV infection and progression of HIV-associated neurocognitive disorders (HAND). However, given the inherent limitations in the existing animal models of HAND, these investigations only recapitulated specific aspects of the disease but not the complex human syndrome. Despite the inability of HIV to infect rodents over the last 30 years, multiple strategies have been employed to develop several rodent models of HAND. While none of these models can accurately mimic the overall pathophysiology of HAND, they serve the purpose of modeling some unique aspects of HAND. This review provides an overview of various animal models used in the field and a careful evaluation of methodological strengths and limitations inherent in both the model systems and study designs to understand better how the various animal models complement one another.
Collapse
Affiliation(s)
- Susmita Sil
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Annadurai Thangaraj
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Ernest T Chivero
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Fang Niu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Muthukumar Kannan
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Ke Liao
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Peter S Silverstein
- School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, 64108, USA
| | - Palsamy Periyasamy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| | - Shilpa Buch
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
24
|
Barbour AJ, Nass SR, Hahn YK, Hauser KF, Knapp PE. Restoration of KCC2 Membrane Localization in Striatal Dopamine D2 Receptor-Expressing Medium Spiny Neurons Rescues Locomotor Deficits in HIV Tat-Transgenic Mice. ASN Neuro 2021; 13:17590914211022089. [PMID: 34445881 PMCID: PMC8404672 DOI: 10.1177/17590914211022089] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 04/23/2021] [Accepted: 05/06/2021] [Indexed: 11/17/2022] Open
Abstract
People infected with HIV (PWH) are highly susceptible to striatal and hippocampal damage. Motor and memory impairments are common among these patients, likely as behavioral manifestations of damage to these brain regions. GABAergic dysfunction from HIV infection and viral proteins such as transactivator of transcription (Tat) have been well documented. We recently demonstrated that the neuron specific Cl- extruder, K+ Cl- cotransporter 2 (KCC2), is diminished after exposure to HIV proteins, including Tat, resulting in disrupted GABAAR-mediated hyperpolarization and inhibition. Here, we utilized doxycycline (DOX)-inducible, GFAP-driven HIV-1 Tat transgenic mice to further explore this phenomenon. After two weeks of Tat expression, we found no changes in hippocampal KCC2 levels, but a significant decrease in the striatum that was associated with hyperlocomotion in the open field assay. We were able to restore KCC2 activity and baseline locomotion with the KCC2 enhancer, CLP290. Additionally, we found that CLP290, whose mechanism of action has yet to be described, acts to restore phosphorylation of serine 940 resulting in increased KCC2 membrane localization. We also examined neuronal subpopulation contributions to the noted effects and found significant differences. Dopamine D2 receptor-expressing medium spiny neurons (MSNs) were selectively vulnerable to Tat-induced KCC2 loss, with no changes observed in dopamine D1 receptor-expressing MSNs. These results suggest that disinhibition/diminished hyperpolarization of dopamine D2 receptor-expressing MSNs can manifest as increased locomotion in this context. They further suggest that KCC2 activity might be a therapeutic target to alleviate motor disturbances related to HIV.
Collapse
Affiliation(s)
- Aaron J. Barbour
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, United States
| | - Sara R. Nass
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, United States
| | - Yun K. Hahn
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, United States
| | - Kurt F. Hauser
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, United States
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, United States
- Institute for Drug and Alcohol Studies, Virginia Commonwealth University School of Medicine, Richmond, Virginia, United States
| | - Pamela E. Knapp
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, United States
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, United States
- Institute for Drug and Alcohol Studies, Virginia Commonwealth University School of Medicine, Richmond, Virginia, United States
| |
Collapse
|
25
|
Chang L, Liang H, Kandel SR, He JJ. Independent and Combined Effects of Nicotine or Chronic Tobacco Smoking and HIV on the Brain: A Review of Preclinical and Clinical Studies. J Neuroimmune Pharmacol 2020; 15:658-693. [PMID: 33108618 DOI: 10.1007/s11481-020-09963-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 09/25/2020] [Indexed: 02/07/2023]
Abstract
Tobacco smoking is highly prevalent among HIV-infected individuals. Chronic smokers with HIV showed greater cognitive deficits and impulsivity, and had more psychopathological symptoms and greater neuroinflammation than HIV non-smokers or smokers without HIV infection. However, preclinical studies that evaluated the combined effects of HIV-infection and tobacco smoking are scare. The preclinical models typically used cell cultures or animal models that involved specific HIV viral proteins or the administration of nicotine to rodents. These preclinical models consistently demonstrated that nicotine had neuroprotective and anti-inflammatory effects, leading to cognitive enhancement. Although the major addictive ingredient in tobacco smoking is nicotine, chronic smoking does not lead to improved cognitive function in humans. Therefore, preclinical studies designed to unravel the interactive effects of chronic tobacco smoking and HIV infection are needed. In this review, we summarized the preclinical studies that demonstrated the neuroprotective effects of nicotine, the neurotoxic effects of the HIV viral proteins, and the scant literature on nicotine or tobacco smoke in HIV transgenic rat models. We also reviewed the clinical studies that evaluated the neurotoxic effects of tobacco smoking, HIV infection and their combined effects on the brain, including studies that evaluated the cognitive and behavioral assessments, as well as neuroimaging measures. Lastly, we compared the different approaches between preclinical and clinical studies, identified some gaps and proposed some future directions. Graphical abstract Independent and combined effects of HIV and tobacco/nicotine. Left top and bottom panels: Both clinical studies of HIV infected persons and preclinical studies using viral proteins in vitro or in vivo in animal models showed that HIV infection could lead to neurotoxicity and neuroinflammation. Right top and bottom panels: While clinical studies of tobacco smoking consistently showed deleterious effects of smoking, clinical and preclinical studies that used nicotine show mild cognitive enhancement, neuroprotective and possibly anti-inflammatory effects. In the developing brain, however, nicotine is neurotoxic. Middle overlapping panels: Clinical studies of persons with HIV who were smokers typically showed additive deleterious effects of HIV and tobacco smoking. However, in the preclinical studies, when nicotine was administered to the HIV-1 Tg rats, the neurotoxic effects of HIV were attenuated, but tobacco smoke worsened the inflammatory cascade.
Collapse
Affiliation(s)
- Linda Chang
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, 670 W. Baltimore Street, HSF III, Baltimore, MD, 21201, USA.
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Medicine, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, USA.
| | - Huajun Liang
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, 670 W. Baltimore Street, HSF III, Baltimore, MD, 21201, USA
| | - Suresh R Kandel
- Department of Microbiology and Immunology, Chicago Medical School, Center for Cancer Cell Biology, Immunology and Infection, Rosalind Franklin University, 3333 Green Bay Road, Basic Science Building 2.300, North Chicago, IL, 60064, USA
| | - Johnny J He
- Department of Microbiology and Immunology, Chicago Medical School, Center for Cancer Cell Biology, Immunology and Infection, Rosalind Franklin University, 3333 Green Bay Road, Basic Science Building 2.300, North Chicago, IL, 60064, USA.
| |
Collapse
|
26
|
Sivalingam K, Cirino TJ, McLaughlin JP, Samikkannu T. HIV-Tat and Cocaine Impact Brain Energy Metabolism: Redox Modification and Mitochondrial Biogenesis Influence NRF Transcription-Mediated Neurodegeneration. Mol Neurobiol 2020; 58:490-504. [PMID: 32978730 DOI: 10.1007/s12035-020-02131-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 09/14/2020] [Indexed: 02/07/2023]
Abstract
HIV infection and drugs of abuse induce oxidative stress and redox imbalance, which cause neurodegeneration. The mechanisms by which HIV infection and cocaine consumption affect astrocyte energy metabolism, and how this leads to neurodegenerative dysfunction, remain poorly understood. Presently, we investigated how oxidative injury causes the depletion of energy resources and glutathione synthetase (GSS), which in turn activates 5' AMP-activated protein kinase (AMPK), glycolytic enzymes, and mitochondrial biogenesis, finally resulting in nuclear factor erythroid (NRF) transcription in astrocytes. Both human primary astrocytes incubated with HIV-1 Tat protein in vitro and HIV-inducible Tat (iTat) mice exposed to cocaine showed decreased levels of GSS and increased superoxide dismutase (SOD) levels. These changes, in turn, significantly activated AMPK and raised the concentrations of several glycolytic enzymes, along with oxidative phosphorylation, the mitochondrial biogenesis of peroxisome proliferator-activated receptor-γ coactivator (PGC-1α) and mitochondrial transcription factor (TFAM), and Nrf1 and Nrf2 gene transcription and protein expression. Moreover, neurons exposed to HIV-1Tat/cocaine-conditioned media showed reductions in dendritic formation, spine density, and neuroplasticity compared with control neurons. These results suggest that redox inhibition of GSS altered AMPK activation and mitochondrial biogenesis to influence Nrf transcription. These processes are important components of the astrocyte signaling network regulating brain energy metabolism in HIV-positive cocaine users. In conclusion, HIV-1 Tat alters redox inhibition, thus increasing glycolytic metabolic profiles and mitochondrial biogenesis, leading to Nrf transcription, and ultimately impacting astrocyte energy resource and metabolism. Cocaine exacerbated these effects, leading to a worsening of neurodegeneration.
Collapse
Affiliation(s)
- Kalaiselvi Sivalingam
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University, 1010 W Avenue B, Kingsville, TX, 78363, USA
| | - Thomas J Cirino
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32611, USA
| | - Jay P McLaughlin
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32611, USA
| | - Thangavel Samikkannu
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University, 1010 W Avenue B, Kingsville, TX, 78363, USA.
| |
Collapse
|
27
|
Baek EJ, Kim H, Basova LA, Rosander A, Kesby JP, Semenova S, Marcondes MCG. Sex differences and Tat expression affect dopaminergic receptor expression and response to antioxidant treatment in methamphetamine-sensitized HIV Tat transgenic mice. Neuropharmacology 2020; 178:108245. [PMID: 32783894 DOI: 10.1016/j.neuropharm.2020.108245] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 06/11/2020] [Accepted: 07/14/2020] [Indexed: 02/07/2023]
Abstract
Methamphetamine (Meth) abuse is a common HIV comorbidity. Males and females differ in their patterns of Meth use, associated behaviors, and responses, but the underlying mechanisms and impact of HIV infection are unclear. Transgenic mice with inducible HIV-1 Tat protein in the brain (iTat) replicate many neurological aspects of HIV infection in humans. We previously showed that Tat induction enhances the Meth sensitization response associated with perturbation of the dopaminergic system, in male iTat mice. Here, we used the iTat mouse model to investigate sex differences in individual and interactive effects of Tat and Meth challenge on locomotor sensitization, brain expression of dopamine receptors (DRDs) and regulatory adenosine receptors (ADORAs). Because Meth administration increases the production of reactive oxygen species (ROS), we also determined whether the effects of Meth could be rescued by concomitant treatment with the ROS scavenger N-acetyl cysteine (NAC). After Meth sensitization and a 7-day abstinence period, groups of Tat+ and Tat-male and female mice were challenged with Meth in combination with NAC. We confirmed that Tat expression and Meth challenge suppressed DRD mRNA and protein in males and females' brains, and showed that females were particularly susceptible to the effects of Meth on D1-like and D2-like DRD subtypes and ADORAs. The expression of these markers differed strikingly between males and females, and between females in different phases of the estrous cycle, in a Tat -dependent manner. NAC attenuated Meth-induced locomotor sensitization and preserved DRD expression in all groups except for Tat + females. These data identify complex interactions between sex, Meth use, and HIV infection on addiction responses, with potential implications for the treatment of male and female Meth users in the context of HIV, especially those with cognitive disorders.
Collapse
Affiliation(s)
- Eun Ji Baek
- The Scripps Research Institute, Neurosciences Department, La Jolla, CA, 92037, USA; Queensland Brain Institute, University of Queensland, St. Lucia, QLD, 4072, Australia
| | - Hahoon Kim
- The Scripps Research Institute, Neurosciences Department, La Jolla, CA, 92037, USA; Queensland Brain Institute, University of Queensland, St. Lucia, QLD, 4072, Australia
| | - Liana A Basova
- San Diego Biomedical Research Institute, San Diego, CA, 92121, USA; The Scripps Research Institute, Neurosciences Department, La Jolla, CA, 92037, USA
| | - Ashley Rosander
- San Diego Biomedical Research Institute, San Diego, CA, 92121, USA
| | - James P Kesby
- Queensland Brain Institute, University of Queensland, St. Lucia, QLD, 4072, Australia; Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, QLD, 4029, Australia; Department of Psychiatry, School of Medicine, University of California San Diego, La Jolla, CA, 92037, USA
| | - Svetlana Semenova
- Department of Psychiatry, School of Medicine, University of California San Diego, La Jolla, CA, 92037, USA
| | - Maria Cecilia Garibaldi Marcondes
- San Diego Biomedical Research Institute, San Diego, CA, 92121, USA; The Scripps Research Institute, Neurosciences Department, La Jolla, CA, 92037, USA.
| |
Collapse
|
28
|
Salahuddin MF, Qrareya AN, Mahdi F, Jackson D, Foster M, Vujanovic T, Box JG, Paris JJ. Combined HIV-1 Tat and oxycodone activate the hypothalamic-pituitary-adrenal and -gonadal axes and promote psychomotor, affective, and cognitive dysfunction in female mice. Horm Behav 2020; 119:104649. [PMID: 31821792 PMCID: PMC7071558 DOI: 10.1016/j.yhbeh.2019.104649] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 09/26/2019] [Accepted: 11/26/2019] [Indexed: 12/31/2022]
Abstract
The majority of HIV+ patients present with neuroendocrine dysfunction and ~50% experience co-morbid neurological symptoms including motor, affective, and cognitive dysfunction, collectively termed neuroHIV. In preclinical models, the neurotoxic HIV-1 regulatory protein, trans-activator of transcription (Tat), promotes neuroHIV pathology that can be exacerbated by opioids. We and others find gonadal steroids, estradiol (E2) or progesterone (P4), to rescue Tat-mediated pathology. However, the combined effects of Tat and opioids on neuroendocrine function and the subsequent ameliorative capacity of gonadal steroids are unknown. We found that conditional HIV-1 Tat expression in naturally-cycling transgenic mice dose-dependently potentiated oxycodone-mediated psychomotor behavior. Tat increased depression-like behavior in a tail-suspension test among proestrous mice, but decreased it among diestrous mice (who already demonstrated greater depression-like behavior); oxycodone reversed these effects. Combined Tat and oxycodone produced apparent behavioral disinhibition of anxiety-like responding which was greater on diestrus than on proestrus. These mice made more central entries in an open field, but spent less time there and demonstrated greater circulating corticosterone. Tat increased the E2:P4 ratio of circulating steroids on diestrus and acute oxycodone attenuated this effect, but repeated oxycodone exacerbated it. Corticotropin-releasing factor was increased by Tat expression, acute oxycodone exposure, and was greater on diestrus compared to proestrus. In human neuroblastoma cells, Tat exerted neurotoxicity that was ameliorated by E2 (1 or 10 nM) or P4 (100, but not 10 nM) independent of oxycodone. Oxycodone decreased gene expression of estrogen and κ-opioid receptors. Thus, neuroendocrine function may be an important target for HIV-1 Tat/opioid interactions.
Collapse
Affiliation(s)
- Mohammed F Salahuddin
- Department of BioMolecular Sciences, University of Mississippi, School of Pharmacy, University, MS 38677-1848, USA
| | - Alaa N Qrareya
- Department of BioMolecular Sciences, University of Mississippi, School of Pharmacy, University, MS 38677-1848, USA
| | - Fakhri Mahdi
- Department of BioMolecular Sciences, University of Mississippi, School of Pharmacy, University, MS 38677-1848, USA
| | - Dejun Jackson
- Department of BioMolecular Sciences, University of Mississippi, School of Pharmacy, University, MS 38677-1848, USA
| | - Matthew Foster
- Department of BioMolecular Sciences, University of Mississippi, School of Pharmacy, University, MS 38677-1848, USA
| | - Tamara Vujanovic
- Department of BioMolecular Sciences, University of Mississippi, School of Pharmacy, University, MS 38677-1848, USA
| | - J Gaston Box
- Department of BioMolecular Sciences, University of Mississippi, School of Pharmacy, University, MS 38677-1848, USA
| | - Jason J Paris
- Department of BioMolecular Sciences, University of Mississippi, School of Pharmacy, University, MS 38677-1848, USA; Research Institute of Pharmaceutical Sciences, University of Mississippi, University, MS 38677, USA.
| |
Collapse
|
29
|
Cirino TJ, Harden SW, McLaughlin JP, Frazier CJ. Region-specific effects of HIV-1 Tat on intrinsic electrophysiological properties of pyramidal neurons in mouse prefrontal cortex and hippocampus. J Neurophysiol 2020; 123:1332-1341. [PMID: 32101482 DOI: 10.1152/jn.00029.2020] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Human immunodeficiency virus (HIV)-1 transactivator of transcription protein (Tat) is a viral protein that promotes transcription of the HIV genome and possesses cell-signaling properties. Long-term exposure of central nervous system (CNS) tissue to HIV-1 Tat is theorized to contribute to HIV-associated neurodegenerative disorder (HAND). In the current study, we sought to directly evaluate the effect of HIV-1 Tat expression on the intrinsic electrophysiological properties of pyramidal neurons located in layer 2/3 of the medial prefrontal cortex and in area CA1 of the hippocampus. Toward that end, we drove Tat expression with doxycycline (100 mg·kg-1·day-1 ip) in inducible Tat (iTat) transgenic mice for 7 days and then performed single-cell electrophysiological studies in acute tissue slices made through the prefrontal cortex and hippocampus. Control experiments were performed in doxycycline-treated G-tg mice, which retain the tetracycline-sensitive promoter but do not express Tat. Our results indicated that the predominant effects of HIV-1 Tat expression are excitatory in medial prefrontal cortical pyramidal neurons yet inhibitory in hippocampal pyramidal neurons. Notably, in these two populations, HIV-1 Tat expression produced differential effects on neuronal gain, membrane time constant, resting membrane potential, and rheobase. Similarly, we also observed distinct effects on action potential kinetics and afterhyperpolarization, as well as on the current-voltage relationship in subthreshold voltage ranges. Collectively, these data provide mechanistic evidence of complex and region-specific changes in neuronal physiology by which HIV-1 Tat protein may promote cognitive deficits associated with HAND.NEW & NOTEWORTHY We drove expression of human immunodeficiency virus (HIV)-1 transactivator of transcription protein (Tat) protein in inducible Tat (iTat) transgenic mice for 7 days and then examined the effects on the intrinsic electrophysiological properties of pyramidal neurons located in the medial prefrontal cortex (mPFC) and in the hippocampus. Our results reveal a variety of specific changes that promote increased intrinsic excitability of layer II/III mPFC pyramidal neurons and decreased intrinsic excitability of hippocampal CA1 pyramidal neurons, highlighting both cell type and region-specific effects.
Collapse
Affiliation(s)
- Thomas J Cirino
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida
| | - Scott W Harden
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida
| | - Jay P McLaughlin
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida
| | - Charles J Frazier
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida
| |
Collapse
|
30
|
Huynh YW, Thompson BM, Larsen CE, Buch S, Guo ML, Bevins RA, Murray JE. Male HIV-1 transgenic rats show reduced cocaine-maintained lever-pressing compared to F344 wildtype rats despite similar baseline locomotion. J Exp Anal Behav 2020; 113:468-484. [PMID: 32077125 DOI: 10.1002/jeab.586] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 01/29/2020] [Accepted: 02/02/2020] [Indexed: 12/12/2022]
Abstract
The HIV-1 transgenic (Tg) rat model is valuable for understanding HIV-associated neurocognitive disorders (HAND) and accompanying substance use and misuse. Tg and F344/NHsd wildtype (WT) rats were allowed to self-administer intrajugular cocaine. For the first 7 sessions, neither genotype self-administered cocaine (0.1 mg/kg/infusion) on a fixed ratio 1 schedule. We thus implemented a lever-cocaine "autoshaping" session followed by a series of manipulations changing dose and reinforcement schedule. Tg rats self-administered much less cocaine than WT rats throughout the study. Of 8 Tg rats, 5 modestly increased self-administration from sessions 36-50. Of those, only 3 showed a lever discrimination. Of 10 WT rats, 8 acquired robust self-administration by session 19; all WT rats self-administered cocaine by the end of the study. WT and Tg rats had similar baseline locomotor activity in the self-administration chamber suggesting that the low levels of cocaine intake in the Tg rats did not reflect a nonspecific motor impairment in this rat strain. Concomitant measurement of activity with self-administration revealed activity increases that followed increased cocaine intake. That relation held in Tg rats. Therefore, the present study provides evidence that HIV-1 Tg rats are less sensitive to the reinforcing effects of cocaine than their F344 WT counterparts.
Collapse
Affiliation(s)
- Y Wendy Huynh
- Department of Psychology, University of Nebraska-, Lincoln
| | | | | | - Shilpa Buch
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha
| | - Ming-Lei Guo
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha
| | - Rick A Bevins
- Department of Psychology, University of Nebraska-, Lincoln
| | | |
Collapse
|
31
|
Pregnane steroidogenesis is altered by HIV-1 Tat and morphine: Physiological allopregnanolone is protective against neurotoxic and psychomotor effects. Neurobiol Stress 2020; 12:100211. [PMID: 32258256 PMCID: PMC7109513 DOI: 10.1016/j.ynstr.2020.100211] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 01/08/2020] [Accepted: 01/20/2020] [Indexed: 02/07/2023] Open
Abstract
Pregnane steroids, particularly allopregnanolone (AlloP), are neuroprotective in response to central insult. While unexplored in vivo, AlloP may confer protection against the neurological dysfunction associated with human immunodeficiency virus type 1 (HIV-1). The HIV-1 regulatory protein, trans-activator of transcription (Tat), is neurotoxic and its expression in mice increases anxiety-like behavior; an effect that can be ameliorated by progesterone, but not when 5α-reduction is blocked. Given that Tat's neurotoxic effects involve mitochondrial dysfunction and can be worsened with opioid exposure, we hypothesized that Tat and/or combined morphine would perturb steroidogenesis in mice, promoting neuronal death, and that exogenous AlloP would rescue these effects. Like other models of neural injury, conditionally inducing HIV-1 Tat in transgenic mice significantly increased the central synthesis of pregnenolone and progesterone's 5α-reduced metabolites, including AlloP, while decreasing central deoxycorticosterone (independent of changes in plasma). Morphine significantly increased brain and plasma concentrations of several steroids (including progesterone, deoxycorticosterone, corticosterone, and their metabolites) likely via activation of the hypothalamic-pituitary-adrenal stress axis. Tat, but not morphine, caused glucocorticoid resistance in primary splenocytes. In neurons, Tat depolarized mitochondrial membrane potential and increased cell death. Physiological concentrations of AlloP (0.1, 1, or 10 nM) reversed these effects. High-concentration AlloP (100 nM) was neurotoxic in combination with morphine. Tat induction in transgenic mice potentiated the psychomotor effects of acute morphine, while exogenous AlloP (1.0 mg/kg, but not 0.5 mg/kg) was ameliorative. Data demonstrate that steroidogenesis is altered by HIV-1 Tat or morphine and that physiological AlloP attenuates resulting neurotoxic and psychomotor effects.
Collapse
|
32
|
Giacometti LL, Barker JM. Comorbid HIV infection and alcohol use disorders: Converging glutamatergic and dopaminergic mechanisms underlying neurocognitive dysfunction. Brain Res 2019; 1723:146390. [PMID: 31421128 PMCID: PMC6766419 DOI: 10.1016/j.brainres.2019.146390] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 08/02/2019] [Accepted: 08/13/2019] [Indexed: 10/26/2022]
Abstract
Alcohol use disorders (AUDs) are highly comorbid with human immunodeficiency virus (HIV) infection, occurring at nearly twice the rate in HIV positive individuals as in the general population. Individuals with HIV who consume alcohol show worse long-term prognoses and may be at elevated risk for the development of HIV-associated neurocognitive disorders. The direction of this relationship is unclear, and likely multifactorial. Chronic alcohol exposure and HIV infection independently promote cognitive dysfunction and further may interact to exacerbate neurocognitive deficits through effects on common targets, including corticostriatal glutamate and dopamine neurotransmission. Additionally, drug and alcohol use is likely to reduce treatment adherence, potentially resulting in accelerated disease progression and subsequent neurocognitive impairment. The development of neurocognitive impairments may further reduce cognitive control over behavior, resulting in escalating alcohol use. This review will examine the complex relationship between HIV infection and alcohol use, highlighting impacts on dopamine and glutamate systems by which alcohol use and HIV act independently and in tandem to alter corticostriatal circuit structure and function to dysregulate cognitive function.
Collapse
Affiliation(s)
- Laura L Giacometti
- Department of Pharmacology and Physiology, Drexel University College of Medicine, United States
| | - Jacqueline M Barker
- Department of Pharmacology and Physiology, Drexel University College of Medicine, United States.
| |
Collapse
|
33
|
Jacobs IR, Xu C, Hermes DJ, League AF, Xu C, Nath B, Jiang W, Niphakis MJ, Cravatt BF, Mackie K, Mukhopadhyay S, Lichtman AH, Ignatowska-Jankowska BM, Fitting S. Inhibitory Control Deficits Associated with Upregulation of CB 1R in the HIV-1 Tat Transgenic Mouse Model of Hand. J Neuroimmune Pharmacol 2019; 14:661-678. [PMID: 31372820 PMCID: PMC6898753 DOI: 10.1007/s11481-019-09867-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 07/11/2019] [Indexed: 12/14/2022]
Abstract
In the era of combined antiretroviral therapy, HIV-1 infected individuals are living longer lives; however, longevity is met with an increasing number of HIV-1 associated neurocognitive disorders (HAND) diagnoses. The transactivator of transcription (Tat) is known to mediate the neurotoxic effects in HAND by acting directly on neurons and also indirectly via its actions on glia. The Go/No-Go (GNG) task was used to examine HAND in the Tat transgenic mouse model. The GNG task involves subjects discriminating between two stimuli sets in order to determine whether or not to inhibit a previously trained response. Data reveal inhibitory control deficits in female Tat(+) mice (p = .048) and an upregulation of cannabinoid type 1 receptors (CB1R) in the infralimbic (IL) cortex in the same female Tat(+) group (p < .05). A significant negative correlation was noted between inhibitory control and IL CB1R expression (r = −.543, p = .045), with CB1R expression predicting 30% of the variance of inhibitory control (R2 = .295, p = .045). Furthermore, there was a significant increase in spontaneous excitatory postsynaptic current (sEPSC) frequencies in Tat(+) compared to Tat(−) mice (p = .008, across sexes). The increase in sEPSC frequency was significantly attenuated by bath application of PF3845, a fatty acid amide hydrolase (FAAH) enzyme inhibitor (p < .001). Overall, the GNG task is a viable measure to assess inhibitory control deficits in Tat transgenic mice and results suggest a potential therapeutic treatment for the observed deficits with drugs which modulate endocannabinoid enzyme activity. Results of the Go/No-Go operant conditioning task reveal inhibitory control deficits in female transgenic Tat(+) mice without significantly affecting males. The demonstrated inhibitory control deficits appear to be associated with an upregulation of cannabinoid type 1 receptors (CB1R) in the infralimbic (IL) cortex in the same female Tat(+) group. ![]()
Collapse
MESH Headings
- AIDS Dementia Complex/genetics
- AIDS Dementia Complex/metabolism
- AIDS Dementia Complex/psychology
- Animals
- Disease Models, Animal
- Female
- HIV-1
- Inhibition, Psychological
- Limbic Lobe/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Neurocognitive Disorders/genetics
- Neurocognitive Disorders/metabolism
- Psychomotor Performance/physiology
- Receptor, Cannabinoid, CB1/biosynthesis
- Receptor, Cannabinoid, CB1/genetics
- Up-Regulation/physiology
- tat Gene Products, Human Immunodeficiency Virus/biosynthesis
- tat Gene Products, Human Immunodeficiency Virus/genetics
Collapse
Affiliation(s)
- Ian R Jacobs
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| | - Changqing Xu
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Douglas J Hermes
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Alexis F League
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Callie Xu
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Bhupendra Nath
- Department of Chemistry & Biochemistry, North Carolina Central University, Durham, NC, 27707, USA
| | - Wei Jiang
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, 29425, USA
- Division of Infectious Diseases, Department of Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Micah J Niphakis
- The Skaggs Institute for Chemical Biology, Department of Chemistry, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Benjamin F Cravatt
- The Skaggs Institute for Chemical Biology, Department of Chemistry, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Ken Mackie
- Department of Psychological & Brain Sciences, Indiana University, Bloomington, IN, 47405, USA
| | - Somnath Mukhopadhyay
- Department of Chemistry & Biochemistry, North Carolina Central University, Durham, NC, 27707, USA
| | - Aron H Lichtman
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | | | - Sylvia Fitting
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
34
|
Mohseni Ahooyi T, Shekarabi M, Decoppet EA, Langford D, Khalili K, Gordon J. Network analysis of hippocampal neurons by microelectrode array in the presence of HIV-1 Tat and cocaine. J Cell Physiol 2018; 233:9299-9311. [PMID: 29206302 DOI: 10.1002/jcp.26322] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 11/27/2017] [Indexed: 12/18/2022]
Abstract
HIV-associated neurocognitive disorders affecting greater than 30% of patients are caused by HIV-1 infection of the CNS, and in part, include neurotoxic effects of the viral transactivator of transcription, Tat protein. In addition to increasing the risk for becoming HIV infected, cocaine abuse enhances the neuropathogenic impacts of HIV-1. To investigate the outcome of Tat and cocaine interference in the hippocampal neuronal network, cross-rank-corrlation was employed to develop a systematic framework to assess hippocampal neurons behavior cultured on multielectrode arrays. Tat and cocaine differentially disturbed neuronal spiking rates, amplitude, synchronous activity, and oscillations within the hippocampal neuronal network via potentiation of inhibitory neurotransmission. The Tat-mediated impairment of neuronal spiking was reversible by removal of Tat, which restored neuronal activity. The presence of astrocytes co-cultured with neuronal networks diminished the effects of Tat and cocaine on neuron function suggesting a role for astrocytes in stabilizing neuronal behavior and increasing neuronal spontaneous activities such as bursting amplitude, frequency, and wave propagation rate. Taken together, our studies indicate that the HIV protein Tat and cocaine impair hippocampal neuronal network functioning and that the presence of astrocytes alleviates network dysfunction pointing to a newly discovered pathway through which ionic homeostasis is maintained by neuron-glial crosstalk in the CNS.
Collapse
Affiliation(s)
- Taha Mohseni Ahooyi
- Department of Neuroscience, Center for Neurovirology and Comprehensive NeuroAIDS Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Masoud Shekarabi
- Department of Neuroscience, Center for Neurovirology and Comprehensive NeuroAIDS Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Emilie A Decoppet
- Department of Neuroscience, Center for Neurovirology and Comprehensive NeuroAIDS Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Dianne Langford
- Department of Neuroscience, Center for Neurovirology and Comprehensive NeuroAIDS Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Kamel Khalili
- Department of Neuroscience, Center for Neurovirology and Comprehensive NeuroAIDS Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Jennifer Gordon
- Department of Neuroscience, Center for Neurovirology and Comprehensive NeuroAIDS Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| |
Collapse
|
35
|
Ohene-Nyako M, Persons AL, Napier TC. Region-specific changes in markers of neuroplasticity revealed in HIV-1 transgenic rats by low-dose methamphetamine. Brain Struct Funct 2018; 223:3503-3513. [PMID: 29931627 DOI: 10.1007/s00429-018-1701-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 06/15/2018] [Indexed: 12/15/2022]
Abstract
Methamphetamine abuse co-occurring with HIV infection presents neuropathology in brain regions that mediate reward and motivation. A neuronal signaling cascade altered acutely by meth and some HIV-1 proteins is the mitogen-activated protein kinase (MAPK) pathway. It remains unknown if chronic co-exposure to meth and HIV-1 proteins converge on MAPK in vivo. To make this determination, we studied young adult Fischer 344 HIV-1 transgenic (Tg) and non-Tg rats that self-administered meth (0.02-0.04 mg/kg/0.05 ml iv infusion, 2 h/day for 21 days) and their saline-yoked controls. One day following the operant task, rats were killed. Brain regions involved in reward-motivation [i.e., nucleus accumbens (NA) and ventral pallidum (VP)], were assayed for a MAPK cascade protein, extracellular signal-regulated kinase (ERK), and a downstream transcription factor, ΔFosB. In the NA, activated (phosphorylated; p) ERK-to-ERK ratio (pERK/ERK) was increased in meth-exposed Tg rats versus saline Tg controls, and versus meth non-Tg rats. ΔFosB was increased in meth Tg rats versus saline and meth non-Tg rats. Assessment of two targets of ΔFosB-regulated transcription revealed (1) increased dopamine D1 receptor (D1R) immunoreactivity in the NA shell of Tg-meth rats versus saline Tg controls, but (2) no changes in the AMPA receptor subunit, GluA2. No changes related to genotype or meth occurred for ERK, ΔFosB or D1R protein in the VP. Results reveal a region-specific activation of ERK, and increases in ΔFosB and D1R expression induced by HIV-1 proteins and meth. Such effects may contribute to the neuronal and behavioral pathology associated with meth/HIV comorbidity.
Collapse
Affiliation(s)
- Michael Ohene-Nyako
- Department of Pharmacology, Rush University, Chicago, IL, USA.,Center for Compulsive Behavior and Addiction, Rush University, Chicago, IL, USA
| | - Amanda L Persons
- Department of Physician Assistant Studies, Rush University, Chicago, IL, USA.,Department of Psychiatry, Rush University Medical Center, 1735 W. Harrison Street, Cohn Research Building Suite #424, Chicago, IL, 60612, USA.,Center for Compulsive Behavior and Addiction, Rush University, Chicago, IL, USA
| | - T Celeste Napier
- Department of Psychiatry, Rush University Medical Center, 1735 W. Harrison Street, Cohn Research Building Suite #424, Chicago, IL, 60612, USA. .,Center for Compulsive Behavior and Addiction, Rush University, Chicago, IL, USA.
| |
Collapse
|
36
|
HIV-1 proteins dysregulate motivational processes and dopamine circuitry. Sci Rep 2018; 8:7869. [PMID: 29777165 PMCID: PMC5959859 DOI: 10.1038/s41598-018-25109-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 04/16/2018] [Indexed: 12/22/2022] Open
Abstract
Motivational alterations, such as apathy, in HIV-1+ individuals are associated with decreased performance on tasks involving frontal-subcortical circuitry. We used the HIV-1 transgenic (Tg) rat to assess effect of long-term HIV-1 protein exposure on motivated behavior using sucrose (1–30%, w/v) and cocaine (0.01–1.0 mg/kg/infusion) maintained responding with fixed-ratio (FR) and progressive-ratio (PR) schedules of reinforcement. For sucrose-reinforced responding, HIV-1 Tg rats displayed no change in EC50 relative to controls, suggesting no change in sucrose reinforcement but had a downward shifted concentration-response curves, suggesting a decrease in response vigor. Cocaine-maintained responding was attenuated in HIV-1 Tg rats (FR1 0.33 mg/kg/infusion and PR 1.0 mg/kg/infusion). Dose-response tests (PR) revealed that HIV-1 Tg animals responded significantly less than F344 control rats and failed to earn significantly more infusions of cocaine as the unit dose increased. When choosing between cocaine and sucrose, control rats initially chose sucrose but with time shifted to a cocaine preference. In contrast, HIV-1 disrupted choice behaviors. DAT function was altered in the striatum of HIV-1 Tg rats; however, prior cocaine self-administration produced a unique effect on dopamine homeostasis in the HIV-1 Tg striatum. These findings of altered goal directed behaviors may determine neurobiological mechanisms of apathy in HIV-1+ patients.
Collapse
|
37
|
Gonek M, McLane VD, Stevens DL, Lippold K, Akbarali HI, Knapp PE, Dewey WL, Hauser KF, Paris JJ. CCR5 mediates HIV-1 Tat-induced neuroinflammation and influences morphine tolerance, dependence, and reward. Brain Behav Immun 2018; 69:124-138. [PMID: 29146238 PMCID: PMC5857418 DOI: 10.1016/j.bbi.2017.11.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 10/02/2017] [Accepted: 11/07/2017] [Indexed: 12/16/2022] Open
Abstract
The HIV-1 regulatory protein, trans-activator of transcription (Tat), interacts with opioids to potentiate neuroinflammation and neurodegeneration within the CNS. These effects may involve the C-C chemokine receptor type 5 (CCR5); however, the behavioral contribution of CCR5 on Tat/opioid interactions is not known. Using a transgenic murine model that expresses HIV-1 Tat protein in a GFAP-regulated, doxycycline-inducible manner, we assessed morphine tolerance, dependence, and reward. To assess the influence of CCR5 on these effects, mice were pretreated with oral vehicle or the CCR5 antagonist, maraviroc, prior to morphine administration. We found that HIV-1 Tat expression significantly attenuated the antinociceptive potency of acute morphine (2-64 mg/kg, i.p.) in non-tolerant mice. Consistent with this, Tat attenuated withdrawal symptoms among morphine-tolerant mice. Pretreatment with maraviroc blocked the effects of Tat, reinstating morphine potency in non-tolerant mice and restoring withdrawal symptomology in morphine-tolerant mice. Twenty-four hours following morphine administration, HIV-1 Tat significantly potentiated (∼3.5-fold) morphine-conditioned place preference and maraviroc further potentiated these effects (∼5.7-fold). Maraviroc exerted no measurable behavioral effects on its own. Protein array analyses revealed only minor changes to cytokine profiles when morphine was administered acutely or repeatedly; however, 24 h post morphine administration, the expression of several cytokines was greatly increased, including endogenous CCR5 chemokine ligands (CCL3, CCL4, and CCL5), as well as CCL2. Tat further elevated levels of several cytokines and maraviroc pretreatment attenuated these effects. These data demonstrate that CCR5 mediates key aspects of HIV-1 Tat-induced alterations in the antinociceptive potency and rewarding properties of opioids.
Collapse
Affiliation(s)
- Maciej Gonek
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, P.O. Box 980613, VA 23298-0613, USA
| | - Virginia D. McLane
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, P.O. Box 980613, VA 23298-0613, USA
| | - David L. Stevens
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, P.O. Box 980613, VA 23298-0613, USA
| | - Kumiko Lippold
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, P.O. Box 980613, VA 23298-0613, USA
| | - Hamid I. Akbarali
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, P.O. Box 980613, VA 23298-0613, USA
| | - Pamela E. Knapp
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, P.O. Box 980613, VA 23298-0613, USA,Department of Anatomy and Neurobiology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, P.O. Box 980709, Richmond, VA 23298-0709, USA,Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, P.O. Box 980059, Richmond, VA 23298-0059, USA
| | - William L. Dewey
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, P.O. Box 980613, VA 23298-0613, USA,Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, P.O. Box 980059, Richmond, VA 23298-0059, USA
| | - Kurt F. Hauser
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, P.O. Box 980613, VA 23298-0613, USA,Department of Anatomy and Neurobiology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, P.O. Box 980709, Richmond, VA 23298-0709, USA,Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, P.O. Box 980059, Richmond, VA 23298-0059, USA
| | - Jason J. Paris
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, P.O. Box 980613, VA 23298-0613, USA,Department of BioMolecular Sciences, University of Mississippi, School of Pharmacy, P.O. Box 1848, University, MS 38677-1848, USA,Research Institute of Pharmaceutical Sciences, University of Mississippi, School of Pharmacy, P.O. Box 1848, University, MS 38677-1848, USA,Address for Correspondence: Jason J. Paris, Ph.D. Assistant Professor of Pharmacology, The University of Mississippi, School of Pharmacy, P.O. Box 1848, 315 Faser Hall, University, MS 38677-1848, U.S.A. Phone: +1-662-915-3096,
| |
Collapse
|
38
|
Persons AL, Bradaric BD, Dodiya HB, Ohene-Nyako M, Forsyth CB, Keshavarzian A, Shaikh M, Napier TC. Colon dysregulation in methamphetamine self-administering HIV-1 transgenic rats. PLoS One 2018; 13:e0190078. [PMID: 29293553 PMCID: PMC5749763 DOI: 10.1371/journal.pone.0190078] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 12/07/2017] [Indexed: 02/07/2023] Open
Abstract
The integrity and function of the gut is impaired in HIV-infected individuals, and gut pathogenesis may play a role in several HIV-associated disorders. Methamphetamine is a popular illicit drug abused by HIV-infected individuals. However, the effect of methamphetamine on the gut and its potential to exacerbate HIV-associated gut pathology is not known. To shed light on this scenario, we evaluated colon barrier pathology in a rat model of the human comorbid condition. Intestinal barrier integrity and permeability were assessed in drug-naïve Fischer 344 HIV-1 transgenic (Tg) and non-Tg rats, and in Tg and non-Tg rats instrumented with jugular cannulae trained to self-administer methamphetamine or serving as saline-yoked controls. Intestinal permeability was determined by measuring the urine content of orally gavaged sugars. Intestinal barrier integrity was evaluated by immunoblotting or immunofluorescence of colon claudin-1 and zonula occludens-1 (ZO-1), two major tight junction proteins that regulate gut epithelial paracellular permeability. Both non-Tg and Tg rats self-administered moderate amounts of methamphetamine. These amounts were sufficient to increase colon permeability, reduce protein level of claudin-1, and reduce claudin-1 and ZO-1 immunofluorescence in Tg rats relative to non-Tg rats. Methamphetamine decreased tight junction immunofluorescence in non-Tg rats, with a similar, but non-significant trend observed in Tg rats. However, the effect of methamphetamine on tight junction proteins was subthreshold to gut leakiness. These findings reveal that both HIV-1 proteins and methamphetamine alter colon barrier integrity, and indicate that the gut may be a pathogenic site for these insults.
Collapse
Affiliation(s)
- Amanda L. Persons
- Department of Psychiatry, Rush University Medical Center, Chicago, IL, United States of America
- Department of Physician Assistant Studies, Rush University Medical Center, Chicago, IL, United States of America
- Center for Compulsive Behavior and Addiction, Rush University Medical Center, Chicago, IL, United States of America
- * E-mail:
| | - Brinda D. Bradaric
- Center for Compulsive Behavior and Addiction, Rush University Medical Center, Chicago, IL, United States of America
- Department of Health Sciences, Rush University Medical Center, Chicago, IL, United States of America
| | - Hemraj B. Dodiya
- Department of Pharmacology, Rush University Medical Center, Chicago, IL, United States of America
| | - Michael Ohene-Nyako
- Center for Compulsive Behavior and Addiction, Rush University Medical Center, Chicago, IL, United States of America
- Department of Pharmacology, Rush University Medical Center, Chicago, IL, United States of America
| | - Christopher B. Forsyth
- Department of Internal Medicine, Division of Digestive Diseases and Nutrition, Rush University Medical Center, Chicago, IL, United States of America
| | - Ali Keshavarzian
- Center for Compulsive Behavior and Addiction, Rush University Medical Center, Chicago, IL, United States of America
- Department of Internal Medicine, Division of Digestive Diseases and Nutrition, Rush University Medical Center, Chicago, IL, United States of America
| | - Maliha Shaikh
- Department of Internal Medicine, Division of Digestive Diseases and Nutrition, Rush University Medical Center, Chicago, IL, United States of America
| | - T. Celeste Napier
- Department of Psychiatry, Rush University Medical Center, Chicago, IL, United States of America
- Center for Compulsive Behavior and Addiction, Rush University Medical Center, Chicago, IL, United States of America
- Department of Pharmacology, Rush University Medical Center, Chicago, IL, United States of America
| |
Collapse
|
39
|
HIV-1 and cocaine disrupt dopamine reuptake and medium spiny neurons in female rat striatum. PLoS One 2017; 12:e0188404. [PMID: 29176843 PMCID: PMC5703481 DOI: 10.1371/journal.pone.0188404] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 11/06/2017] [Indexed: 11/19/2022] Open
Abstract
HIV-1 and addictive drugs, such as cocaine (COC), may act in combination to produce serious neurological complications. In the present experiments, striatal brain slices from HIV-1 transgenic (Tg) and F344 control female rats were studied. First, we examined dopamine (DA) reuptake in control, HIV-1, COC-treated (5µM) and HIV-1+COC-treated, striatal slices using fast scan cyclic voltammetry. COC-treated striatal slices from F344 control animals significantly increased DA reuptake time (T80), relative to untreated control slices. In contrast, in HIV-1 Tg striatal slices, DA reuptake time was extended by HIV-1, which was not further altered by COC treatment. Second, analysis of medium spiny neuronal populations from striatal brain slices found that controls treated with cocaine displayed increases in spine length, whereas cocaine treated HIV-1 slices displayed decreased spine length. Taken together, the current study provides evidence for dysfunction of the dopamine transporter (DAT) in mediating DA reuptake in HIV-1 Tg rats and limited responses to acute COC exposure. Collectively, dysfunction of the DAT reuptake and altered dendritic spine morphology of the MSNs, suggest a functional disruption of the dopamine system within the HIV-1 Tg rat.
Collapse
|
40
|
Langford D, Oh Kim B, Zou W, Fan Y, Rahimain P, Liu Y, He JJ. Doxycycline-inducible and astrocyte-specific HIV-1 Tat transgenic mice (iTat) as an HIV/neuroAIDS model. J Neurovirol 2017; 24:168-179. [PMID: 29143286 DOI: 10.1007/s13365-017-0598-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 10/08/2017] [Accepted: 11/01/2017] [Indexed: 12/18/2022]
Abstract
HIV-1 Tat is known to be neurotoxic and important for HIV/neuroAIDS pathogenesis. However, the overwhelming majority of the studies involved use of recombinant Tat protein. To understand the contributions of Tat protein to HIV/neuroAIDS and the underlying molecular mechanisms of HIV-1 Tat neurotoxicity in the context of a whole organism and independently of HIV-1 infection, a doxycycline-inducible astrocyte-specific HIV-1 Tat transgenic mouse (iTat) was created. Tat expression in the brains of iTat mice was determined to be in the range of 1-5 ng/ml and led to astrocytosis, loss of neuronal dendrites, and neuroinflammation. iTat mice have allowed us to define the direct effects of Tat on astrocytes and the molecular mechanisms of Tat-induced GFAP expression/astrocytosis, astrocyte-mediated Tat neurotoxicity, Tat-impaired neurogenesis, Tat-induced loss of neuronal integrity, and exosome-associated Tat release and uptake. In this review, we will provide an overview about the creation and characterization of this model and its utilities for our understanding of Tat neurotoxicity and the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Dianne Langford
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Byung Oh Kim
- School of Food Science & Biotechnology and College of Agriculture & Life Sciences, Kyungpook National University, Daegu, 702-701, South Korea
| | - Wei Zou
- The 1st Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Yan Fan
- Graduate School of Biomedical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX, 76107, USA
| | - Pejman Rahimain
- Graduate School of Biomedical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX, 76107, USA
| | - Ying Liu
- Graduate School of Biomedical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX, 76107, USA
| | - Johnny J He
- Graduate School of Biomedical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX, 76107, USA.
| |
Collapse
|
41
|
Abstract
HIV-associated neurocognitive disorder (HAND) remains highly prevalent in HIV infected individuals and represents a special group of neuropathological disorders, which are associated with HIV-1 viral proteins, such as transactivator of transcription (Tat) protein. Cocaine abuse increases the incidence of HAND and exacerbates its severity by enhancing viral replication. Perturbation of dopaminergic transmission has been implicated as a risk factor of HAND. The presynaptic dopamine (DA) transporter (DAT) is essential for DA homeostasis and dopaminergic modulation of the brain function including cognition. Tat and cocaine synergistically elevate synaptic DA levels by acting directly on human DAT (hDAT), ultimately leading to dysregulation of DA transmission. Through integrated computational modeling and experimental validation, key residues have been identified in hDAT that play a critical role in Tat-induced inhibition of DAT and induce transporter conformational transitions. This review presents current information regarding neurological changes in DAT-mediated dopaminergic system associated with HIV infection, DAT-mediated adaptive responses to Tat as well as allosteric modulatory effects of novel compounds on hDAT. Understanding the molecular mechanisms by which Tat induces DAT-mediated dysregulation of DA system is of great clinical interest for identifying new targets for an early therapeutic intervention for HAND.
Collapse
|
42
|
HIV-1 TAT protein enhances sensitization to methamphetamine by affecting dopaminergic function. Brain Behav Immun 2017; 65:210-221. [PMID: 28495611 PMCID: PMC5537017 DOI: 10.1016/j.bbi.2017.05.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 04/28/2017] [Accepted: 05/06/2017] [Indexed: 12/14/2022] Open
Abstract
Methamphetamine abuse is common among humans with immunodeficiency virus (HIV). The HIV-1 regulatory protein TAT induces dysfunction of mesolimbic dopaminergic systems which may result in impaired reward processes and contribute to methamphetamine abuse. These studies investigated the impact of TAT expression on methamphetamine-induced locomotor sensitization, underlying changes in dopamine function and adenosine receptors in mesolimbic brain areas and neuroinflammation (microgliosis). Transgenic mice with doxycycline-induced TAT protein expression in the brain were tested for locomotor activity in response to repeated methamphetamine injections and methamphetamine challenge after a 7-day abstinence period. Dopamine function in the nucleus accumbens (Acb) was determined using high performance liquid chromatography. Expression of dopamine and/or adenosine A receptors (ADORA) in the Acb and caudate putamen (CPu) was assessed using RT-PCR and immunohistochemistry analyses. Microarrays with pathway analyses assessed dopamine and adenosine signaling in the CPu. Activity-dependent neurotransmitter switching of a reserve pool of non-dopaminergic neurons to a dopaminergic phenotype in the ventral tegmental area (VTA) was determined by immunohistochemistry and quantified with stereology. TAT expression enhanced methamphetamine-induced sensitization. TAT expression alone decreased striatal dopamine (D1, D2, D4, D5) and ADORA1A receptor expression, while increasing ADORA2A receptors expression. Moreover, TAT expression combined with methamphetamine exposure was associated with increased adenosine A receptors (ADORA1A) expression and increased recruitment of dopamine neurons in the VTA. TAT expression and methamphetamine exposure induced microglia activation with the largest effect after combined exposure. Our findings suggest that dopamine-adenosine receptor interactions and reserve pool neuronal recruitment may represent potential targets to develop new treatments for methamphetamine abuse in individuals with HIV.
Collapse
|
43
|
McLaughlin JP, Paris JJ, Mintzopoulos D, Hymel KA, Kim JK, Cirino TJ, Gillis TE, Eans SO, Vitaliano GD, Medina JM, Krapf RC, Stacy HM, Kaufman MJ. Conditional Human Immunodeficiency Virus Transactivator of Transcription Protein Expression Induces Depression-like Effects and Oxidative Stress. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2017; 2:599-609. [PMID: 29057370 PMCID: PMC5648358 DOI: 10.1016/j.bpsc.2017.04.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
BACKGROUND The prevalence of major depression in those with HIV/AIDS is substantially higher than in the general population. Mechanisms underlying this comorbidity are poorly understood. HIV-transactivator of transcription (Tat) protein, produced and excreted by HIV, could be involved. We determined whether conditional Tat protein expression in mice is sufficient to induce depression-like behaviors and oxidative stress. Further, as oxidative stress is associated with depression, we determined whether decreasing or increasing oxidative stress by administering methylsulfonylmethane (MSM) or diethylmaleate (DEM), respectively, altered depression-like behavior. METHODS GT-tg bigenic mice received intraperitoneal saline or doxycycline (Dox, 25-100 mg/kg/day) to induce Tat expression. G-tg mice, which do not express Tat protein, also received Dox. Depression-like behavior was assessed with the tail suspension test (TST) and the two-bottle saccharin/water consumption task. Reactive oxygen/nitrogen species (ROS/RNS) were assessed ex vivo. Medial frontal cortex (MFC) oxidative stress and temperature were measured in vivo with 9.4-Tesla proton magnetic resonance spectroscopy (MRS). RESULTS Tat expression increased TST immobility time in an exposure-dependent manner and reduced saccharin consumption. MSM decreased immobility time while DEM increased it in saline-treated GT-tg mice. Tat and MSM behavioral effects persisted for 28 days. Tat and DEM increased while MSM decreased ROS/RNS levels. Tat expression increased MFC glutathione levels and temperature. CONCLUSIONS Tat expression induced rapid and enduring depression-like behaviors and oxidative stress. Increasing/decreasing oxidative stress increased/decreased, respectively, depression-like behavior. Thus, Tat produced by HIV may contribute to the high depression prevalence among those with HIV. Further, mitigation of oxidative stress could reduce depression severity.
Collapse
Affiliation(s)
- Jay P. McLaughlin
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32610
| | - Jason J. Paris
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32610
- Virginia Commonwealth University, Department of Pharmacology & Toxicology, Richmond, VA 23298
| | - Dionyssios Mintzopoulos
- McLean Imaging Center, Department of Psychiatry, McLean Hospital/Harvard Medical School, Belmont, MA 02478
| | - Kristen A. Hymel
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32610
| | - Jae K. Kim
- McLean Imaging Center, Department of Psychiatry, McLean Hospital/Harvard Medical School, Belmont, MA 02478
| | - Thomas J. Cirino
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32610
| | - Timothy E. Gillis
- McLean Imaging Center, Department of Psychiatry, McLean Hospital/Harvard Medical School, Belmont, MA 02478
| | - Shainnel O. Eans
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32610
| | - Gordana D. Vitaliano
- McLean Imaging Center, Department of Psychiatry, McLean Hospital/Harvard Medical School, Belmont, MA 02478
| | - Jessica M. Medina
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32610
| | - Richard C. Krapf
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32610
| | - Heather M. Stacy
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32610
| | - Marc J. Kaufman
- McLean Imaging Center, Department of Psychiatry, McLean Hospital/Harvard Medical School, Belmont, MA 02478
| |
Collapse
|
44
|
Gaskill PJ, Miller DR, Gamble-George J, Yano H, Khoshbouei H. HIV, Tat and dopamine transmission. Neurobiol Dis 2017; 105:51-73. [PMID: 28457951 PMCID: PMC5541386 DOI: 10.1016/j.nbd.2017.04.015] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 04/04/2017] [Accepted: 04/16/2017] [Indexed: 01/02/2023] Open
Abstract
Human Immunodeficiency Virus (HIV) is a progressive infection that targets the immune system, affecting more than 37 million people around the world. While combinatorial antiretroviral therapy (cART) has lowered mortality rates and improved quality of life in infected individuals, the prevalence of HIV associated neurocognitive disorders is increasing and HIV associated cognitive decline remains prevalent. Recent research has suggested that HIV accessory proteins may be involved in this decline, and several studies have indicated that the HIV protein transactivator of transcription (Tat) can disrupt normal neuronal and glial function. Specifically, data indicate that Tat may directly impact dopaminergic neurotransmission, by modulating the function of the dopamine transporter and specifically damaging dopamine-rich regions of the CNS. HIV infection of the CNS has long been associated with dopaminergic dysfunction, but the mechanisms remain undefined. The specific effect(s) of Tat on dopaminergic neurotransmission may be, at least partially, a mechanism by which HIV infection directly or indirectly induces dopaminergic dysfunction. Therefore, precisely defining the specific effects of Tat on the dopaminergic system will help to elucidate the mechanisms by which HIV infection of the CNS induces neuropsychiatric, neurocognitive and neurological disorders that involve dopaminergic neurotransmission. Further, this will provide a discussion of the experiments needed to further these investigations, and may help to identify or develop new therapeutic approaches for the prevention or treatment of these disorders in HIV-infected individuals.
Collapse
Affiliation(s)
- Peter J Gaskill
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, United States.
| | - Douglas R Miller
- Department of Neuroscience, University of Florida, Gainesville, FL 32611, United States
| | - Joyonna Gamble-George
- Department of Neuroscience, University of Florida, Gainesville, FL 32611, United States
| | - Hideaki Yano
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, United States
| | - Habibeh Khoshbouei
- Department of Neuroscience, University of Florida, Gainesville, FL 32611, United States.
| |
Collapse
|
45
|
HIV Tat Impairs Neurogenesis through Functioning As a Notch Ligand and Activation of Notch Signaling Pathway. J Neurosci 2017; 36:11362-11373. [PMID: 27807176 DOI: 10.1523/jneurosci.1208-16.2016] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 09/10/2016] [Indexed: 02/07/2023] Open
Abstract
Alterations in adult neurogenesis have been noted in the brain of HIV-infected individuals and are likely linked to HIV-associated neurocognitive deficits, including those in learning and memory. But the underlying molecular mechanisms are not fully understood. In the study, we took advantage of doxycycline-inducible and astrocyte-specific HIV-1 Tat transgenic mice (iTat) and determined the relationship between Tat expression and neurogenesis. Tat expression in astrocytes was associated with fewer neuron progenitor cells (NPCs), fewer immature neurons, and fewer mature neurons in the dentate gyrus of the hippocampus of the mouse brain. In vitro NPC-derived neurosphere assays showed that Tat-containing conditioned media from astrocytes or recombinant Tat protein inhibited NPC proliferation and migration and altered NPC differentiation, while immunodepletion of Tat from Tat-containing conditioned media or heat inactivation of recombinant Tat abrogated those effects. Notch signaling downstream gene Hes1 promoter-driven luciferase reporter gene assay and Western blotting showed that recombinant Tat or Tat-containing conditioned media activated Hes1 transcription and protein expression, which were abrogated by Tat heat inactivation, immunodepletion, and cysteine mutation at position 30. Last, Notch signaling inhibitor N-[N-(3,5-difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester (DAPT) significantly rescued Tat-impaired NPC differentiation in vitro and neurogenesis in vivo Together, these results show that Tat adversely affects NPCs and neurogenesis through Notch signaling and point to the potential of developing Notch signaling inhibitors as HIV/neuroAIDS therapeutics. SIGNIFICANCE STATEMENT HIV infection of the CNS causes cognitive and memory deficits, which have become more prevalent in the era of combination antiretroviral therapy (cART). Neurogenesis is impaired in HIV-infected individuals. But the underlying molecular mechanisms remain largely unknown. In this study, we have discovered that HIV Tat impairs neurogenesis through the Notch signaling pathway. These findings are particularly important because Tat protein has recently been detected in the brain of HIV-infected individuals with HIV replication in the periphery being effectively controlled by cART. The current study not only further highlights the importance of HIV Tat protein in HIV/neuroAIDS, but also presents a new strategy to develop novel HIV/neuroAIDS therapeutics, particularly in the era of cART.
Collapse
|
46
|
Sun WL, Quizon PM, Yuan Y, Zhang W, Ananthan S, Zhan CG, Zhu J. Allosteric modulatory effects of SRI-20041 and SRI-30827 on cocaine and HIV-1 Tat protein binding to human dopamine transporter. Sci Rep 2017. [PMID: 28623359 PMCID: PMC5473888 DOI: 10.1038/s41598-017-03771-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Dopamine transporter (DAT) is the target of cocaine and HIV-1 transactivator of transcription (Tat) protein. Identifying allosteric modulatory molecules with potential attenuation of cocaine and Tat binding to DAT are of great scientific and clinical interest. We demonstrated that tyrosine 470 and 88 act as functional recognition residues in human DAT (hDAT) for Tat-induced inhibition of DA transport and transporter conformational transitions. Here we investigated the allosteric modulatory effects of two allosteric ligands, SRI-20041 and SRI-30827 on cocaine binding on wild type (WT) hDAT, Y470 H and Y88 F mutants. Effect of SRI-30827 on Tat-induced inhibition of [3H]WIN35,428 binding was also determined. Compared to a competitive DAT inhibitor indatraline, both SRI-compounds displayed a similar decrease (30%) in IC50 for inhibition of [3H]DA uptake by cocaine in WT hDAT. The addition of SRI-20041 or SRI-30827 following cocaine slowed the dissociation rate of [3H]WIN35,428 binding in WT hDAT relative to cocaine alone. Moreover, Y470H and Y88F hDAT potentiate the inhibitory effect of cocaine on DA uptake and attenuate the effects of SRI-compounds on cocaine-mediated dissociation rate. SRI-30827 attenuated Tat-induced inhibition of [3H]WIN35,428 binding. These observations demonstrate that tyrosine 470 and 88 are critical for allosteric modulatory effects of SRI-compounds on the interaction of cocaine with hDAT.
Collapse
Affiliation(s)
- Wei-Lun Sun
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, USA
| | - Pamela M Quizon
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, USA
| | - Yaxia Yuan
- Molecular Modeling and Biopharmaceutical Center, University of Kentucky, Lexington, KY, USA.,Center for Pharmaceutical Research and Innovation, University of Kentucky, Lexington, KY, USA.,Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, USA
| | - Wei Zhang
- Department of Chemistry, Drug Discovery Division, Southern Research Institute, Birmingham, AL, USA
| | - Subramaniam Ananthan
- Department of Chemistry, Drug Discovery Division, Southern Research Institute, Birmingham, AL, USA
| | - Chang-Guo Zhan
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, USA.,Center for Pharmaceutical Research and Innovation, University of Kentucky, Lexington, KY, USA.,Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, USA
| | - Jun Zhu
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, USA.
| |
Collapse
|
47
|
Molecular mechanism: the human dopamine transporter histidine 547 regulates basal and HIV-1 Tat protein-inhibited dopamine transport. Sci Rep 2016; 6:39048. [PMID: 27966610 PMCID: PMC5155291 DOI: 10.1038/srep39048] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 11/17/2016] [Indexed: 12/15/2022] Open
Abstract
Abnormal dopaminergic transmission has been implicated as a risk determinant of HIV-1-associated neurocognitive disorders. HIV-1 Tat protein increases synaptic dopamine (DA) levels by directly inhibiting DA transporter (DAT) activity, ultimately leading to dopaminergic neuron damage. Through integrated computational modeling prediction and experimental validation, we identified that histidine547 on human DAT (hDAT) is critical for regulation of basal DA uptake and Tat-induced inhibition of DA transport. Compared to wild type hDAT (WT hDAT), mutation of histidine547 (H547A) displayed a 196% increase in DA uptake. Other substitutions of histidine547 showed that DA uptake was not altered in H547R but decreased by 99% in H547P and 60% in H547D, respectively. These mutants did not alter DAT surface expression or surface DAT binding sites. H547 mutants attenuated Tat-induced inhibition of DA transport observed in WT hDAT. H547A displays a differential sensitivity to PMA- or BIM-induced activation or inhibition of DAT function relative to WT hDAT, indicating a change in basal PKC activity in H547A. These findings demonstrate that histidine547 on hDAT plays a crucial role in stabilizing basal DA transport and Tat-DAT interaction. This study provides mechanistic insights into identifying targets on DAT for Tat binding and improving DAT-mediated dysfunction of DA transmission.
Collapse
|
48
|
Kesby JP, Markou A, Semenova S. The effects of HIV-1 regulatory TAT protein expression on brain reward function, response to psychostimulants and delay-dependent memory in mice. Neuropharmacology 2016; 109:205-215. [PMID: 27316905 DOI: 10.1016/j.neuropharm.2016.06.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 06/08/2016] [Accepted: 06/13/2016] [Indexed: 10/21/2022]
Abstract
Depression and psychostimulant abuse are common comorbidities among humans with immunodeficiency virus (HIV) disease. The HIV regulatory protein TAT is one of multiple HIV-related proteins associated with HIV-induced neurotoxicity. TAT-induced dysfunction of dopamine and serotonin systems in corticolimbic brain areas may result in impaired reward function, thus, contributing to depressive symptoms and psychostimulant abuse. Transgenic mice with doxycycline-induced TAT protein expression in the brain (TAT+, TAT- control) show neuropathology resembling brain abnormalities in HIV+ humans. We evaluated brain reward function in response to TAT expression, nicotine and methamphetamine administration in TAT+ and TAT- mice using the intracranial self-stimulation procedure. We evaluated the brain dopamine and serotonin systems with high-performance liquid chromatography. The effects of TAT expression on delay-dependent working memory in TAT+ and TAT- mice using the operant delayed nonmatch-to-position task were also assessed. During doxycycline administration, reward thresholds were elevated by 20% in TAT+ mice compared with TAT- mice. After the termination of doxycycline treatment, thresholds of TAT+ mice remained significantly higher than those of TAT- mice and this was associated with changes in mesolimbic serotonin and dopamine levels. TAT+ mice showed a greater methamphetamine-induced threshold lowering compared with TAT- mice. TAT expression did not alter delay-dependent working memory. These results indicate that TAT expression in mice leads to reward deficits, a core symptom of depression, and a greater sensitivity to methamphetamine-induced reward enhancement. Our findings suggest that the TAT protein may contribute to increased depressive-like symptoms and continued methamphetamine use in HIV-positive individuals.
Collapse
Affiliation(s)
- James P Kesby
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA; Queensland Brain Institute, The University of Queensland, St. Lucia, Qld, Australia
| | - Athina Markou
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Svetlana Semenova
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
49
|
Hahn YK, Paris JJ, Lichtman AH, Hauser KF, Sim-Selley LJ, Selley DE, Knapp PE. 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 β-arrestin 2 activity in the forebrain. Neurobiol Dis 2016; 92:124-36. [PMID: 26845176 DOI: 10.1016/j.nbd.2016.01.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 12/31/2015] [Accepted: 01/19/2016] [Indexed: 02/06/2023] Open
Abstract
Co-exposure to opiates and HIV/HIV proteins results in enhanced CNS morphological and behavioral deficits in HIV(+) individuals and in animal models. Opiates with abuse liability, such as heroin and morphine, bind preferentially to and have pharmacological actions through μ-opioid-receptors (MORs). The mechanisms underlying opiate-HIV interactions are not understood. Exposure to the HIV-1 transactivator of transcription (Tat) protein causes neurodegenerative outcomes that parallel many aspects of the human disease. We have also observed that in vivo exposure to Tat results in apparent changes in morphine efficacy, and thus have hypothesized that HIV proteins might alter MOR activation. To test our hypothesis, MOR-mediated G-protein activation was determined in neuroAIDS-relevant forebrain regions of transgenic mice with inducible CNS expression of HIV-1 Tat. G-protein activation was assessed by MOR agonist-stimulated [(35)S]guanosine-5'-O-(3-thio)triphosphate ([(35)S]GTPγS) autoradiography in brain sections, and in concentration-effect curves of MOR agonist-stimulated [(35)S]GTPγS binding in membranes isolated from specific brain regions. Comparative studies were done using the MOR-selective agonist DAMGO ([D-Ala(2), N-MePhe(4), Gly-ol]-enkephalin) and a more clinically relevant agonist, morphine. Tat exposure reduced MOR-mediated G-protein activation in an agonist, time, and regionally dependent manner. Levels of the GPCR regulatory protein β-arrestin-2, which is involved in MOR desensitization, were found to be elevated in only one affected brain region, the amygdala; amygdalar β-arrestin-2 also showed a significantly increased association with MOR by co-immunoprecipitation, suggesting decreased availability of MOR. Interestingly, this correlated with changes in anxiety and fear-conditioned extinction, behaviors that have substantial amygdalar input. We propose that HIV-1 Tat alters the intrinsic capacity of MOR to signal in response to agonist binding, possibly via a mechanism involving altered expression and/or function of β-arrestin-2.
Collapse
Affiliation(s)
- Yun K Hahn
- Department of Anatomy & Neurobiology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, VA 23298-0709, USA
| | - Jason J Paris
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, VA 23298-0613, USA
| | - Aron H Lichtman
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, VA 23298-0613, USA; Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Richmond, VA 23298-0059, USA
| | - Kurt F Hauser
- Department of Anatomy & Neurobiology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, VA 23298-0709, USA; Department of Pharmacology & Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, VA 23298-0613, USA; Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Richmond, VA 23298-0059, USA
| | - Laura J Sim-Selley
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, VA 23298-0613, USA; Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Richmond, VA 23298-0059, USA
| | - Dana E Selley
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, VA 23298-0613, USA; Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Richmond, VA 23298-0059, USA
| | - Pamela E Knapp
- Department of Anatomy & Neurobiology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, VA 23298-0709, USA; Department of Pharmacology & Toxicology, Virginia Commonwealth University, Medical College of Virginia (MCV) Campus, Richmond, VA 23298-0613, USA; Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Richmond, VA 23298-0059, USA.
| |
Collapse
|
50
|
Maubert ME, Pirrone V, Rivera NT, Wigdahl B, Nonnemacher MR. Interaction between Tat and Drugs of Abuse during HIV-1 Infection and Central Nervous System Disease. Front Microbiol 2016; 6:1512. [PMID: 26793168 PMCID: PMC4707230 DOI: 10.3389/fmicb.2015.01512] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 12/15/2015] [Indexed: 02/02/2023] Open
Abstract
In many individuals, drug abuse is intimately linked with HIV-1 infection. In addition to being associated with one-third of all HIV-1 infections in the United States, drug abuse also plays a role in disease progression and severity in HIV-1-infected patients, including adverse effects on the central nervous system (CNS). Specific systems within the brain are known to be damaged in HIV-1-infected individuals and this damage is similar to that observed in drug abuse. Even in the era of anti-retroviral therapy (ART), CNS pathogenesis occurs with HIV-1 infection, with a broad range of cognitive impairment observed, collectively referred to as HIV-1-associated neurocognitive disorders (HAND). A number of HIV-1 proteins (Tat, gp120, Nef, Vpr) have been implicated in the etiology of pathogenesis and disease as a result of the biologic activity of the extracellular form of each of the proteins in a number of tissues, including the CNS, even in ART-suppressed patients. In this review, we have made Tat the center of attention for a number of reasons. First, it has been shown to be synthesized and secreted by HIV-1-infected cells in the CNS, despite the most effective suppression therapies available to date. Second, Tat has been shown to alter the functions of several host factors, disrupting the molecular and biochemical balance of numerous pathways contributing to cellular toxicity, dysfunction, and death. In addition, the advantages and disadvantages of ART suppression with regard to controlling the genesis and progression of neurocognitive impairment are currently under debate in the field and are yet to be fully determined. In this review, we discuss the individual and concerted contributions of HIV-1 Tat, drug abuse, and ART with respect to damage in the CNS, and how these factors contribute to the development of HAND in HIV-1-infected patients.
Collapse
Affiliation(s)
- Monique E Maubert
- Department of Microbiology and Immunology, Drexel University College of MedicinePhiladelphia, PA, USA; Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of MedicinePhiladelphia, PA, USA
| | - Vanessa Pirrone
- Department of Microbiology and Immunology, Drexel University College of MedicinePhiladelphia, PA, USA; Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of MedicinePhiladelphia, PA, USA
| | - Nina T Rivera
- Department of Microbiology and Immunology, Drexel University College of MedicinePhiladelphia, PA, USA; Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of MedicinePhiladelphia, PA, USA
| | - Brian Wigdahl
- Department of Microbiology and Immunology, Drexel University College of MedicinePhiladelphia, PA, USA; Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of MedicinePhiladelphia, PA, USA
| | - Michael R Nonnemacher
- Department of Microbiology and Immunology, Drexel University College of MedicinePhiladelphia, PA, USA; Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of MedicinePhiladelphia, PA, USA
| |
Collapse
|