Skip to main content
Top
Published in: Journal of Neuroinflammation 1/2018

Open Access 01-12-2018 | Research

A central role for glial CCR5 in directing the neuropathological interactions of HIV-1 Tat and opiates

Authors: Sarah Kim, Yun Kyung Hahn, Elizabeth M Podhaizer, Virginia D McLane, Shiping Zou, Kurt F Hauser, Pamela E Knapp

Published in: Journal of Neuroinflammation | Issue 1/2018

Login to get access

Abstract

Background

The collective cognitive and motor deficits known as HIV-associated neurocognitive disorders (HAND) remain high even among HIV+ individuals whose antiretroviral therapy is optimized. HAND is worsened in the context of opiate abuse. The mechanism of exacerbation remains unclear but likely involves chronic immune activation of glial cells resulting from persistent, low-level exposure to the virus and viral proteins. We tested whether signaling through C-C chemokine receptor type 5 (CCR5) contributes to neurotoxic interactions between HIV-1 transactivator of transcription (Tat) and opiates and explored potential mechanisms.

Methods

Neuronal survival was tracked in neuronal and glial co-cultures over 72 h of treatment with HIV-1 Tat ± morphine using cells from CCR5-deficient and wild-type mice exposed to the CCR5 antagonist maraviroc or exogenously-added BDNF (analyzed by repeated measures ANOVA). Intracellular calcium changes in response to Tat ± morphine ± maraviroc were assessed by ratiometric Fura-2 imaging (analyzed by repeated measures ANOVA). Release of brain-derived neurotrophic factor (BDNF) and its precursor proBDNF from CCR5-deficient and wild-type glia was measured by ELISA (analyzed by two-way ANOVA). Levels of CCR5 and μ-opioid receptor (MOR) were measured by immunoblotting (analyzed by Student’s t test).

Results

HIV-1 Tat induces neurotoxicity, which is greatly exacerbated by morphine in wild-type cultures expressing CCR5. Loss of CCR5 from glia (but not neurons) eliminated neurotoxicity due to Tat and morphine interactions. Unexpectedly, when CCR5 was lost from glia, morphine appeared to entirely protect neurons from Tat-induced toxicity. Maraviroc pre-treatment similarly eliminated neurotoxicity and attenuated neuronal increases in [Ca2+]i caused by Tat ± morphine. proBDNF/BDNF ratios were increased in conditioned media from Tat ± morphine-treated wild-type glia compared to CCR5-deficient glia. Exogenous BDNF treatments mimicked the pro-survival effect of glial CCR5 deficiency against Tat ± morphine.

Conclusions

Our results suggest a critical role for glial CCR5 in mediating neurotoxic effects of HIV-1 Tat and morphine interactions on neurons. A shift in the proBDNF/BDNF ratio that favors neurotrophic support may occur when glial CCR5 signaling is blocked. Some neuroprotection occurred only in the presence of morphine, suggesting that loss of CCR5 may fundamentally change signaling through the MOR in glia.
Literature
2.
go back to reference Yazdanian M. Blood-brain barrier properties of human immunodeficiency virus antiretrovirals. J Pharm Sci. 1999;88:950–4.CrossRefPubMed Yazdanian M. Blood-brain barrier properties of human immunodeficiency virus antiretrovirals. J Pharm Sci. 1999;88:950–4.CrossRefPubMed
3.
go back to reference Powderly WG. Current approaches to treatment for HIV-1 infection. J Neuro-Oncol. 2000;6(Suppl 1):S8–s13. Powderly WG. Current approaches to treatment for HIV-1 infection. J Neuro-Oncol. 2000;6(Suppl 1):S8–s13.
4.
go back to reference Resnick L, Berger JR, Shapshak P, Tourtellotte WW. Early penetration of the blood-brain-barrier by HIV. Neurology. 1988;38:9–14.CrossRefPubMed Resnick L, Berger JR, Shapshak P, Tourtellotte WW. Early penetration of the blood-brain-barrier by HIV. Neurology. 1988;38:9–14.CrossRefPubMed
5.
go back to reference Sturdevant CB, Joseph SB, Schnell G, Price RW, Swanstrom R, Spudich S. Compartmentalized replication of R5 T cell-tropic HIV-1 in the central nervous system early in the course of infection. PLoS Pathog. 2015;11:e1004720.CrossRefPubMedCentralPubMed Sturdevant CB, Joseph SB, Schnell G, Price RW, Swanstrom R, Spudich S. Compartmentalized replication of R5 T cell-tropic HIV-1 in the central nervous system early in the course of infection. PLoS Pathog. 2015;11:e1004720.CrossRefPubMedCentralPubMed
6.
go back to reference Xiao Q, Li J, Yu Q, Bao R, Liu J, Liu H, Zhou L, Xian Q, Wang Y, Cheng-Mayer C, et al. Distinct compartmentalization in the CNS of SHIVKU-1-infected Chinese rhesus macaque is associated with severe neuropathology. J Acquir Immune Defic Syndr. 2015;70:e168–71.CrossRefPubMed Xiao Q, Li J, Yu Q, Bao R, Liu J, Liu H, Zhou L, Xian Q, Wang Y, Cheng-Mayer C, et al. Distinct compartmentalization in the CNS of SHIVKU-1-infected Chinese rhesus macaque is associated with severe neuropathology. J Acquir Immune Defic Syndr. 2015;70:e168–71.CrossRefPubMed
7.
go back to reference Bednar MM, Sturdevant CB, Tompkins LA, Arrildt KT, Dukhovlinova E, Kincer LP, Swanstrom R. Compartmentalization, viral evolution, and viral latency of HIV in the CNS. Curr HIV/AIDS Rep. 2015;12:262–71.CrossRefPubMedCentralPubMed Bednar MM, Sturdevant CB, Tompkins LA, Arrildt KT, Dukhovlinova E, Kincer LP, Swanstrom R. Compartmentalization, viral evolution, and viral latency of HIV in the CNS. Curr HIV/AIDS Rep. 2015;12:262–71.CrossRefPubMedCentralPubMed
8.
go back to reference Heaton RK, Clifford DB, Franklin DR Jr, Woods SP, Ake C, Vaida F, Ellis RJ, Letendre SL, Marcotte TD, Atkinson JH, et al. HIV-associated neurocognitive disorders persist in the era of potent antiretroviral therapy: CHARTER study. Neurology. 2010;75:2087–96.CrossRefPubMedCentralPubMed Heaton RK, Clifford DB, Franklin DR Jr, Woods SP, Ake C, Vaida F, Ellis RJ, Letendre SL, Marcotte TD, Atkinson JH, et al. HIV-associated neurocognitive disorders persist in the era of potent antiretroviral therapy: CHARTER study. Neurology. 2010;75:2087–96.CrossRefPubMedCentralPubMed
9.
go back to reference Maschke M, Kastrup O, Esser S, Ross B, Hengge U, Hufnagel A. Incidence and prevalence of neurological disorders associated with HIV since the introduction of highly active antiretroviral therapy (HAART). J Neurol Neurosurg Psychiatry. 2000;69:376–80.CrossRefPubMedCentralPubMed Maschke M, Kastrup O, Esser S, Ross B, Hengge U, Hufnagel A. Incidence and prevalence of neurological disorders associated with HIV since the introduction of highly active antiretroviral therapy (HAART). J Neurol Neurosurg Psychiatry. 2000;69:376–80.CrossRefPubMedCentralPubMed
10.
go back to reference Sacktor N, McDermott MP, Marder K, Schifitto G, Selnes OA, McArthur JC, Stern Y, Albert S, Palumbo D, Kieburtz K, et al. HIV-associated cognitive impairment before and after the advent of combination therapy. J Neuro-Oncol. 2002;8:136–42. Sacktor N, McDermott MP, Marder K, Schifitto G, Selnes OA, McArthur JC, Stern Y, Albert S, Palumbo D, Kieburtz K, et al. HIV-associated cognitive impairment before and after the advent of combination therapy. J Neuro-Oncol. 2002;8:136–42.
11.
go back to reference Antinori A, Arendt G, Becker JT, Brew BJ, Byrd DA, Cherner M, Clifford DB, Cinque P, Epstein LG, Goodkin K, et al. Updated research nosology for HIV-associated neurocognitive disorders. Neurology. 2007;69:1789–99.CrossRefPubMed Antinori A, Arendt G, Becker JT, Brew BJ, Byrd DA, Cherner M, Clifford DB, Cinque P, Epstein LG, Goodkin K, et al. Updated research nosology for HIV-associated neurocognitive disorders. Neurology. 2007;69:1789–99.CrossRefPubMed
12.
go back to reference Bell JE. An update on the neuropathology of HIV in the HAART era. Histopathology. 2004;45:549–59.CrossRefPubMed Bell JE. An update on the neuropathology of HIV in the HAART era. Histopathology. 2004;45:549–59.CrossRefPubMed
13.
go back to reference Everall I, Vaida F, Khanlou N, Lazzaretto D, Achim C, Letendre S, Moore D, Ellis R, Cherner M, Gelman B, et al. Cliniconeuropathologic correlates of human immunodeficiency virus in the era of antiretroviral therapy. J Neuro-Oncol. 2009;15:360–70. Everall I, Vaida F, Khanlou N, Lazzaretto D, Achim C, Letendre S, Moore D, Ellis R, Cherner M, Gelman B, et al. Cliniconeuropathologic correlates of human immunodeficiency virus in the era of antiretroviral therapy. J Neuro-Oncol. 2009;15:360–70.
14.
go back to reference An SF, Groves M, Giometto B, Beckett AA, Scaravilli F. Detection and localisation of HIV-1 DNA and RNA in fixed adult AIDS brain by polymerase chain reaction/in situ hybridisation technique. Acta Neuropathol. 1999;98:481–7.CrossRefPubMed An SF, Groves M, Giometto B, Beckett AA, Scaravilli F. Detection and localisation of HIV-1 DNA and RNA in fixed adult AIDS brain by polymerase chain reaction/in situ hybridisation technique. Acta Neuropathol. 1999;98:481–7.CrossRefPubMed
15.
go back to reference Takahashi K, Wesselingh SL, Griffin DE, McArthur JC, Johnson RT, Glass JD. Localization of HIV-1 in human brain using polymerase chain reaction/in situ hybridization and immunocytochemistry. Ann Neurol. 1996;39:705–11.CrossRefPubMed Takahashi K, Wesselingh SL, Griffin DE, McArthur JC, Johnson RT, Glass JD. Localization of HIV-1 in human brain using polymerase chain reaction/in situ hybridization and immunocytochemistry. Ann Neurol. 1996;39:705–11.CrossRefPubMed
16.
go back to reference Wiley CA, Schrier RD, Nelson JA, Lampert PW, Oldstone MB. Cellular localization of human immunodeficiency virus infection within the brains of acquired immune deficiency syndrome patients. Proc Natl Acad Sci U S A. 1986;83:7089–93.CrossRefPubMedCentralPubMed Wiley CA, Schrier RD, Nelson JA, Lampert PW, Oldstone MB. Cellular localization of human immunodeficiency virus infection within the brains of acquired immune deficiency syndrome patients. Proc Natl Acad Sci U S A. 1986;83:7089–93.CrossRefPubMedCentralPubMed
17.
go back to reference Cosenza MA, Zhao ML, Si Q, Lee SC. Human brain parenchymal microglia express CD14 and CD45 and are productively infected by HIV-1 in HIV-1 encephalitis. Brain Pathol. 2002;12:442–55.CrossRefPubMed Cosenza MA, Zhao ML, Si Q, Lee SC. Human brain parenchymal microglia express CD14 and CD45 and are productively infected by HIV-1 in HIV-1 encephalitis. Brain Pathol. 2002;12:442–55.CrossRefPubMed
18.
go back to reference Kramer-Hammerle S, Rothenaigner I, Wolff H, Bell JE, Brack-Werner R. Cells of the central nervous system as targets and reservoirs of the human immunodeficiency virus. Virus Res. 2005;111:194–213.CrossRefPubMed Kramer-Hammerle S, Rothenaigner I, Wolff H, Bell JE, Brack-Werner R. Cells of the central nervous system as targets and reservoirs of the human immunodeficiency virus. Virus Res. 2005;111:194–213.CrossRefPubMed
19.
go back to reference Williams KC, Hickey WF. Central nervous system damage, monocytes and macrophages, and neurological disorders in AIDS. Annu Rev Neurosci. 2002;25:537–62.CrossRefPubMed Williams KC, Hickey WF. Central nervous system damage, monocytes and macrophages, and neurological disorders in AIDS. Annu Rev Neurosci. 2002;25:537–62.CrossRefPubMed
20.
go back to reference Churchill MJ, Wesselingh SL, Cowley D, Pardo CA, McArthur JC, Brew BJ, Gorry PR. Extensive astrocyte infection is prominent in human immunodeficiency virus-associated dementia. Ann Neurol. 2009;66:253–8.CrossRefPubMed Churchill MJ, Wesselingh SL, Cowley D, Pardo CA, McArthur JC, Brew BJ, Gorry PR. Extensive astrocyte infection is prominent in human immunodeficiency virus-associated dementia. Ann Neurol. 2009;66:253–8.CrossRefPubMed
21.
go back to reference Bagasra O, Lavi E, Bobroski L, Khalili K, Pestaner JP, Tawadros R, Pomerantz RJ. Cellular reservoirs of HIV-1 in the central nervous system of infected individuals: identification by the combination of in situ polymerase chain reaction and immunohistochemistry. AIDS. 1996;10:573–85.CrossRefPubMed Bagasra O, Lavi E, Bobroski L, Khalili K, Pestaner JP, Tawadros R, Pomerantz RJ. Cellular reservoirs of HIV-1 in the central nervous system of infected individuals: identification by the combination of in situ polymerase chain reaction and immunohistochemistry. AIDS. 1996;10:573–85.CrossRefPubMed
22.
go back to reference Anthony IC, Arango JC, Stephens B, Simmonds P, Bell JE. The effects of illicit drugs on the HIV infected brain. Front Biosci. 2008;13:1294–307.CrossRefPubMed Anthony IC, Arango JC, Stephens B, Simmonds P, Bell JE. The effects of illicit drugs on the HIV infected brain. Front Biosci. 2008;13:1294–307.CrossRefPubMed
23.
go back to reference Byrd DA, Fellows RP, Morgello S, Franklin D, Heaton RK, Deutsch R, Atkinson JH, Clifford DB, Collier AC, Marra CM, et al. Neurocognitive impact of substance use in HIV infection. J Acquir Immune Defic Syndr. 2011;58:154–62.CrossRefPubMedCentralPubMed Byrd DA, Fellows RP, Morgello S, Franklin D, Heaton RK, Deutsch R, Atkinson JH, Clifford DB, Collier AC, Marra CM, et al. Neurocognitive impact of substance use in HIV infection. J Acquir Immune Defic Syndr. 2011;58:154–62.CrossRefPubMedCentralPubMed
24.
go back to reference Martin-Thormeyer EM, Paul RH. Drug abuse and hepatitis C infection as comorbid features of HIV associated neurocognitive disorder: neurocognitive and neuroimaging features. Neuropsychol Rev. 2009;19:215–31.CrossRefPubMedCentralPubMed Martin-Thormeyer EM, Paul RH. Drug abuse and hepatitis C infection as comorbid features of HIV associated neurocognitive disorder: neurocognitive and neuroimaging features. Neuropsychol Rev. 2009;19:215–31.CrossRefPubMedCentralPubMed
25.
go back to reference Bell JE, Arango JC, Anthony IC. Neurobiology of multiple insults: HIV-1-associated brain disorders in those who use illicit drugs. J NeuroImmune Pharmacol. 2006;1:182–91.CrossRefPubMed Bell JE, Arango JC, Anthony IC. Neurobiology of multiple insults: HIV-1-associated brain disorders in those who use illicit drugs. J NeuroImmune Pharmacol. 2006;1:182–91.CrossRefPubMed
26.
go back to reference Rogers TJ, Peterson PK. Opioid G protein-coupled receptors: signals at the crossroads of inflammation. Trends Immunol. 2003;24:116–21.CrossRefPubMed Rogers TJ, Peterson PK. Opioid G protein-coupled receptors: signals at the crossroads of inflammation. Trends Immunol. 2003;24:116–21.CrossRefPubMed
27.
go back to reference El-Hage N, Gurwell JA, Singh IN, Knapp PE, Nath A, Hauser KF. Synergistic increases in intracellular Ca2+, and the release of MCP-1, RANTES, and IL-6 by astrocytes treated with opiates and HIV-1 Tat. Glia. 2005;50:91–106.CrossRefPubMedCentralPubMed El-Hage N, Gurwell JA, Singh IN, Knapp PE, Nath A, Hauser KF. Synergistic increases in intracellular Ca2+, and the release of MCP-1, RANTES, and IL-6 by astrocytes treated with opiates and HIV-1 Tat. Glia. 2005;50:91–106.CrossRefPubMedCentralPubMed
28.
go back to reference El-Hage N, Wu G, Wang J, Ambati J, Knapp PE, Reed JL, Bruce-Keller AJ, Hauser KF. HIV-1 Tat and opiate-induced changes in astrocytes promote chemotaxis of microglia through the expression of MCP-1 and alternative chemokines. Glia. 2006;53:132–46.CrossRefPubMedCentralPubMed El-Hage N, Wu G, Wang J, Ambati J, Knapp PE, Reed JL, Bruce-Keller AJ, Hauser KF. HIV-1 Tat and opiate-induced changes in astrocytes promote chemotaxis of microglia through the expression of MCP-1 and alternative chemokines. Glia. 2006;53:132–46.CrossRefPubMedCentralPubMed
29.
go back to reference El-Hage N, Bruce-Keller AJ, Yakovleva T, Bazov I, Bakalkin G, Knapp PE, Hauser KF. Morphine exacerbates HIV-1 Tat-induced cytokine production in astrocytes through convergent effects on [Ca2+]i, NF-κB trafficking and transcription. PLoS One. 2008;3:e4093.CrossRefPubMedCentralPubMed El-Hage N, Bruce-Keller AJ, Yakovleva T, Bazov I, Bakalkin G, Knapp PE, Hauser KF. Morphine exacerbates HIV-1 Tat-induced cytokine production in astrocytes through convergent effects on [Ca2+]i, NF-κB trafficking and transcription. PLoS One. 2008;3:e4093.CrossRefPubMedCentralPubMed
30.
go back to reference Zou S, Fitting S, Hahn YK, Welch SP, El-Hage N, Hauser KF, Knapp PE. Morphine potentiates neurodegenerative effects of HIV-1 Tat through actions at μ-opioid receptor-expressing glia. Brain. 2011;134:3616–31.CrossRefPubMed Zou S, Fitting S, Hahn YK, Welch SP, El-Hage N, Hauser KF, Knapp PE. Morphine potentiates neurodegenerative effects of HIV-1 Tat through actions at μ-opioid receptor-expressing glia. Brain. 2011;134:3616–31.CrossRefPubMed
31.
go back to reference Sorce S, Myburgh R, Krause KH. The chemokine receptor CCR5 in the central nervous system. Prog Neurobiol. 2011;93:297–311.CrossRefPubMed Sorce S, Myburgh R, Krause KH. The chemokine receptor CCR5 in the central nervous system. Prog Neurobiol. 2011;93:297–311.CrossRefPubMed
32.
go back to reference Kelder W, McArthur JC, Nance-Sproson T, McClernon D, Griffin DE. β-chemokines MCP-1 and RANTES are selectively increased in cerebrospinal fluid of patients with human immunodeficiency virus-associated dementia. Ann Neurol. 1998;44:831–5.CrossRefPubMed Kelder W, McArthur JC, Nance-Sproson T, McClernon D, Griffin DE. β-chemokines MCP-1 and RANTES are selectively increased in cerebrospinal fluid of patients with human immunodeficiency virus-associated dementia. Ann Neurol. 1998;44:831–5.CrossRefPubMed
33.
go back to reference McManus CM, Weidenheim K, Woodman SE, Nunez J, Hesselgesser J, Nath A, Berman JW. Chemokine and chemokine-receptor expression in human glial elements: induction by the HIV protein, Tat, and chemokine autoregulation. Am J Pathol. 2000;156:1441–53.CrossRefPubMedCentralPubMed McManus CM, Weidenheim K, Woodman SE, Nunez J, Hesselgesser J, Nath A, Berman JW. Chemokine and chemokine-receptor expression in human glial elements: induction by the HIV protein, Tat, and chemokine autoregulation. Am J Pathol. 2000;156:1441–53.CrossRefPubMedCentralPubMed
34.
go back to reference Martin-Blondel G, Brassat D, Bauer J, Lassmann H, Liblau RS. CCR5 blockade for neuroinflammatory diseases--beyond control of HIV. Nat Rev Neurol. 2016;12:95–105.CrossRefPubMed Martin-Blondel G, Brassat D, Bauer J, Lassmann H, Liblau RS. CCR5 blockade for neuroinflammatory diseases--beyond control of HIV. Nat Rev Neurol. 2016;12:95–105.CrossRefPubMed
35.
go back to reference Dragic T, Litwin V, Allaway GP, Martin SR, Huang Y, Nagashima KA, Cayanan C, Maddon PJ, Koup RA, Moore JP, Paxton WA. HIV-1 entry into CD4+ cells is mediated by the chemokine receptor CC-CKR-5. Nature. 1996;381:667–73.CrossRefPubMed Dragic T, Litwin V, Allaway GP, Martin SR, Huang Y, Nagashima KA, Cayanan C, Maddon PJ, Koup RA, Moore JP, Paxton WA. HIV-1 entry into CD4+ cells is mediated by the chemokine receptor CC-CKR-5. Nature. 1996;381:667–73.CrossRefPubMed
36.
go back to reference Dean M, Carrington M, Winkler C, Huttley GA, Smith MW, Allikmets R, Goedert JJ, Buchbinder SP, Vittinghoff E, Gomperts E, et al. Genetic restriction of HIV-1 infection and progression to AIDS by a deletion allele of the CKR5 structural gene. Hemophilia growth and development study, multicenter AIDS cohort study, multicenter hemophilia cohort study, San Francisco City cohort, ALIVE study. Science. 1996;273:1856–62.CrossRefPubMed Dean M, Carrington M, Winkler C, Huttley GA, Smith MW, Allikmets R, Goedert JJ, Buchbinder SP, Vittinghoff E, Gomperts E, et al. Genetic restriction of HIV-1 infection and progression to AIDS by a deletion allele of the CKR5 structural gene. Hemophilia growth and development study, multicenter AIDS cohort study, multicenter hemophilia cohort study, San Francisco City cohort, ALIVE study. Science. 1996;273:1856–62.CrossRefPubMed
37.
go back to reference Ioannidis JP, Rosenberg PS, Goedert JJ, Ashton LJ, Benfield TL, Buchbinder SP, Coutinho RA, Eugen-Olsen J, Gallart T, Katzenstein TL, et al. Effects of CCR5-Δ32, CCR2-64I, and SDF-1 3’A alleles on HIV-1 disease progression: an international meta-analysis of individual-patient data. Ann Intern Med. 2001;135:782–95.CrossRefPubMed Ioannidis JP, Rosenberg PS, Goedert JJ, Ashton LJ, Benfield TL, Buchbinder SP, Coutinho RA, Eugen-Olsen J, Gallart T, Katzenstein TL, et al. Effects of CCR5-Δ32, CCR2-64I, and SDF-1 3’A alleles on HIV-1 disease progression: an international meta-analysis of individual-patient data. Ann Intern Med. 2001;135:782–95.CrossRefPubMed
38.
go back to reference Levine AJ, Singer EJ, Shapshak P. The role of host genetics in the susceptibility for HIV-associated neurocognitive disorders. AIDS Behav. 2009;13:118–32.CrossRefPubMed Levine AJ, Singer EJ, Shapshak P. The role of host genetics in the susceptibility for HIV-associated neurocognitive disorders. AIDS Behav. 2009;13:118–32.CrossRefPubMed
39.
go back to reference Llibre JM, Rivero A, Rojas JF, Garcia Del Toro M, Herrero C, Arroyo D, Pineda JA, Pasquau J, Masia M, Crespo M, et al. Safety, efficacy and indications of prescription of maraviroc in clinical practice: factors associated with clinical outcomes. Antivir Res. 2015;120:79–84.CrossRefPubMed Llibre JM, Rivero A, Rojas JF, Garcia Del Toro M, Herrero C, Arroyo D, Pineda JA, Pasquau J, Masia M, Crespo M, et al. Safety, efficacy and indications of prescription of maraviroc in clinical practice: factors associated with clinical outcomes. Antivir Res. 2015;120:79–84.CrossRefPubMed
40.
go back to reference Kelly KM, Beck SE, Metcalf Pate KA, Queen SE, Dorsey JL, Adams RJ, Avery LB, Hubbard W, Tarwater PM, Mankowski JL. Neuroprotective maraviroc monotherapy in simian immunodeficiency virus-infected macaques: reduced replicating and latent SIV in the brain. AIDS. 2013;27:F21–8.CrossRefPubMed Kelly KM, Beck SE, Metcalf Pate KA, Queen SE, Dorsey JL, Adams RJ, Avery LB, Hubbard W, Tarwater PM, Mankowski JL. Neuroprotective maraviroc monotherapy in simian immunodeficiency virus-infected macaques: reduced replicating and latent SIV in the brain. AIDS. 2013;27:F21–8.CrossRefPubMed
41.
go back to reference Gates TM, Cysique LA, Siefried KJ, Chaganti J, Moffat KJ, Brew BJ. Maraviroc-intensified combined antiretroviral therapy improves cognition in virally suppressed HIV-associated neurocognitive disorder. AIDS. 2016;30:591–600.CrossRefPubMed Gates TM, Cysique LA, Siefried KJ, Chaganti J, Moffat KJ, Brew BJ. Maraviroc-intensified combined antiretroviral therapy improves cognition in virally suppressed HIV-associated neurocognitive disorder. AIDS. 2016;30:591–600.CrossRefPubMed
42.
go back to reference Ndhlovu LC, Umaki T, Chew GM, Chow DC, Agsalda M, Kallianpur KJ, Paul R, Zhang G, Ho E, Hanks N, et al. Treatment intensification with maraviroc (CCR5 antagonist) leads to declines in CD16-expressing monocytes in cART-suppressed chronic HIV-infected subjects and is associated with improvements in neurocognitive test performance: implications for HIV-associated neurocognitive disease (HAND). J Neuro-Oncol. 2014;20:571–82. Ndhlovu LC, Umaki T, Chew GM, Chow DC, Agsalda M, Kallianpur KJ, Paul R, Zhang G, Ho E, Hanks N, et al. Treatment intensification with maraviroc (CCR5 antagonist) leads to declines in CD16-expressing monocytes in cART-suppressed chronic HIV-infected subjects and is associated with improvements in neurocognitive test performance: implications for HIV-associated neurocognitive disease (HAND). J Neuro-Oncol. 2014;20:571–82.
43.
go back to reference Guo CJ, Li Y, Tian S, Wang X, Douglas SD, Ho WZ. Morphine enhances HIV infection of human blood mononuclear phagocytes through modulation of β-chemokines and CCR5 receptor. J Investig Med. 2002;50:435–42.CrossRefPubMedCentralPubMed Guo CJ, Li Y, Tian S, Wang X, Douglas SD, Ho WZ. Morphine enhances HIV infection of human blood mononuclear phagocytes through modulation of β-chemokines and CCR5 receptor. J Investig Med. 2002;50:435–42.CrossRefPubMedCentralPubMed
44.
go back to reference Miyagi T, Chuang LF, Doi RH, Carlos MP, Torres JV, Chuang RY. Morphine induces gene expression of CCR5 in human CEMx174 lymphocytes. J Biol Chem. 2000;275:31305–10.CrossRefPubMed Miyagi T, Chuang LF, Doi RH, Carlos MP, Torres JV, Chuang RY. Morphine induces gene expression of CCR5 in human CEMx174 lymphocytes. J Biol Chem. 2000;275:31305–10.CrossRefPubMed
45.
go back to reference El-Hage N, Bruce-Keller AJ, Knapp PE, Hauser KF. CCL5/RANTES gene deletion attenuates opioid-induced increases in glial CCL2/MCP-1 immunoreactivity and activation in HIV-1 Tat-exposed mice. J NeuroImmune Pharmacol. 2008;3:275–85.CrossRefPubMedCentralPubMed El-Hage N, Bruce-Keller AJ, Knapp PE, Hauser KF. CCL5/RANTES gene deletion attenuates opioid-induced increases in glial CCL2/MCP-1 immunoreactivity and activation in HIV-1 Tat-exposed mice. J NeuroImmune Pharmacol. 2008;3:275–85.CrossRefPubMedCentralPubMed
46.
go back to reference Avdoshina V, Garzino-Demo A, Bachis A, Monaco MC, Maki PM, Tractenberg RE, Liu C, Young MA, Mocchetti I. HIV-1 decreases the levels of neurotrophins in human lymphocytes. AIDS. 2011;25:1126–8.CrossRefPubMed Avdoshina V, Garzino-Demo A, Bachis A, Monaco MC, Maki PM, Tractenberg RE, Liu C, Young MA, Mocchetti I. HIV-1 decreases the levels of neurotrophins in human lymphocytes. AIDS. 2011;25:1126–8.CrossRefPubMed
47.
go back to reference Bachis A, Avdoshina V, Zecca L, Parsadanian M, Mocchetti I. Human immunodeficiency virus type 1 alters brain-derived neurotrophic factor processing in neurons. J Neurosci. 2012;32:9477–84.CrossRefPubMedCentralPubMed Bachis A, Avdoshina V, Zecca L, Parsadanian M, Mocchetti I. Human immunodeficiency virus type 1 alters brain-derived neurotrophic factor processing in neurons. J Neurosci. 2012;32:9477–84.CrossRefPubMedCentralPubMed
48.
go back to reference Masvekar RR, El-Hage N, Hauser KF, Knapp PE. Morphine enhances HIV-1SF162-mediated neuron death and delays recovery of injured neurites. PLoS One. 2014;9:e100196.CrossRefPubMedCentralPubMed Masvekar RR, El-Hage N, Hauser KF, Knapp PE. Morphine enhances HIV-1SF162-mediated neuron death and delays recovery of injured neurites. PLoS One. 2014;9:e100196.CrossRefPubMedCentralPubMed
49.
go back to reference Fitting S, Knapp PE, Zou S, Marks WD, Bowers MS, Akbarali HI, Hauser KF. Interactive HIV-1 Tat and morphine-induced synaptodendritic injury is triggered through focal disruptions in Na+ influx, mitochondrial instability, and Ca2+ overload. J Neurosci. 2014;34:12850–64.CrossRefPubMed Fitting S, Knapp PE, Zou S, Marks WD, Bowers MS, Akbarali HI, Hauser KF. Interactive HIV-1 Tat and morphine-induced synaptodendritic injury is triggered through focal disruptions in Na+ influx, mitochondrial instability, and Ca2+ overload. J Neurosci. 2014;34:12850–64.CrossRefPubMed
50.
go back to reference Nath A, Conant K, Chen P, Scott C, Major EO. Transient exposure to HIV-1 Tat protein results in cytokine production in macrophages and astrocytes. A hit and run phenomenon. J Biol Chem. 1999;274:17098–102.CrossRefPubMed Nath A, Conant K, Chen P, Scott C, Major EO. Transient exposure to HIV-1 Tat protein results in cytokine production in macrophages and astrocytes. A hit and run phenomenon. J Biol Chem. 1999;274:17098–102.CrossRefPubMed
51.
go back to reference Kruman II, Nath A, Mattson MP. HIV-1 protein Tat induces apoptosis of hippocampal neurons by a mechanism involving caspase activation, calcium overload, and oxidative stress. Exp Neurol. 1998;154:276–88.CrossRefPubMed Kruman II, Nath A, Mattson MP. HIV-1 protein Tat induces apoptosis of hippocampal neurons by a mechanism involving caspase activation, calcium overload, and oxidative stress. Exp Neurol. 1998;154:276–88.CrossRefPubMed
52.
go back to reference Bruce-Keller AJ, Turchan-Cholewo J, Smart EJ, Geurin T, Chauhan A, Reid R, Xu R, Nath A, Knapp PE, Hauser KF. Morphine causes rapid increases in glial activation and neuronal injury in the striatum of inducible HIV-1 Tat transgenic mice. Glia. 2008;56:1414–27.CrossRefPubMedCentralPubMed Bruce-Keller AJ, Turchan-Cholewo J, Smart EJ, Geurin T, Chauhan A, Reid R, Xu R, Nath A, Knapp PE, Hauser KF. Morphine causes rapid increases in glial activation and neuronal injury in the striatum of inducible HIV-1 Tat transgenic mice. Glia. 2008;56:1414–27.CrossRefPubMedCentralPubMed
53.
go back to reference Wiley CA, Baldwin M, Achim CL. Expression of HIV regulatory and structural mRNA in the central nervous system. AIDS. 1996;10:843–7.CrossRefPubMed Wiley CA, Baldwin M, Achim CL. Expression of HIV regulatory and structural mRNA in the central nervous system. AIDS. 1996;10:843–7.CrossRefPubMed
54.
go back to reference Weiss JM, Nath A, Major EO, Berman JW. HIV-1 Tat induces monocyte chemoattractant protein-1-mediated monocyte transmigration across a model of the human blood-brain barrier and up-regulates CCR5 expression on human monocytes. J Immunol. 1999;163:2953–9.PubMed Weiss JM, Nath A, Major EO, Berman JW. HIV-1 Tat induces monocyte chemoattractant protein-1-mediated monocyte transmigration across a model of the human blood-brain barrier and up-regulates CCR5 expression on human monocytes. J Immunol. 1999;163:2953–9.PubMed
55.
go back to reference Ostrowski MA, Justement SJ, Catanzaro A, Hallahan CA, Ehler LA, Mizell SB, Kumar PN, Mican JA, Chun TW, Fauci AS. Expression of chemokine receptors CXCR4 and CCR5 in HIV-1-infected and uninfected individuals. J Immunol. 1998;161:3195–201.PubMed Ostrowski MA, Justement SJ, Catanzaro A, Hallahan CA, Ehler LA, Mizell SB, Kumar PN, Mican JA, Chun TW, Fauci AS. Expression of chemokine receptors CXCR4 and CCR5 in HIV-1-infected and uninfected individuals. J Immunol. 1998;161:3195–201.PubMed
56.
go back to reference Bokhari SM, Yao H, Bethel-Brown C, Fuwang P, Williams R, Dhillon NK, Hegde R, Kumar A, Buch SJ. Morphine enhances Tat-induced activation in murine microglia. J Neuro-Oncol. 2009;15:219–28. Bokhari SM, Yao H, Bethel-Brown C, Fuwang P, Williams R, Dhillon NK, Hegde R, Kumar A, Buch SJ. Morphine enhances Tat-induced activation in murine microglia. J Neuro-Oncol. 2009;15:219–28.
57.
go back to reference 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;69:124–38.CrossRefPubMed 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;69:124–38.CrossRefPubMed
58.
go back to reference Maung R, Hoefer MM, Sanchez AB, Sejbuk NE, Medders KE, Desai MK, Catalan IC, Dowling CC, de Rozieres CM, Garden GA, et al. CCR5 knockout prevents neuronal injury and behavioral impairment induced in a transgenic mouse model by a CXCR4-using HIV-1 glycoprotein 120. J Immunol. 2014;193:1895–910.CrossRefPubMed Maung R, Hoefer MM, Sanchez AB, Sejbuk NE, Medders KE, Desai MK, Catalan IC, Dowling CC, de Rozieres CM, Garden GA, et al. CCR5 knockout prevents neuronal injury and behavioral impairment induced in a transgenic mouse model by a CXCR4-using HIV-1 glycoprotein 120. J Immunol. 2014;193:1895–910.CrossRefPubMed
59.
go back to reference Sun S, Chen D, Lin F, Chen M, Yu H, Hou L, Li C. Role of interleukin-4, the chemokine CCL3 and its receptor CCR5 in neuropathic pain. Mol Immunol. 2016;77:184–92.CrossRefPubMed Sun S, Chen D, Lin F, Chen M, Yu H, Hou L, Li C. Role of interleukin-4, the chemokine CCL3 and its receptor CCR5 in neuropathic pain. Mol Immunol. 2016;77:184–92.CrossRefPubMed
60.
go back to reference Victoria ECG, de Brito Toscano EC, de Sousa Cardoso AC, da Silva DG, de Miranda AS, da Silva Barcelos L, Sugimoto MA, Sousa LP, de Assis Lima IV, de Oliveira ACP, et al. Knockdown of C-C chemokine receptor 5 (CCR5) is protective against cerebral ischemia and reperfusion injury. Curr Neurovasc Res. 2017;14:125–31.CrossRefPubMed Victoria ECG, de Brito Toscano EC, de Sousa Cardoso AC, da Silva DG, de Miranda AS, da Silva Barcelos L, Sugimoto MA, Sousa LP, de Assis Lima IV, de Oliveira ACP, et al. Knockdown of C-C chemokine receptor 5 (CCR5) is protective against cerebral ischemia and reperfusion injury. Curr Neurovasc Res. 2017;14:125–31.CrossRefPubMed
61.
go back to reference Barber TJ, Imaz A, Boffito M, Niubo J, Pozniak A, Fortuny R, Alonso J, Davies N, Mandalia S, Podzamczer D, Gazzard B. CSF inflammatory markers and neurocognitive function after addition of maraviroc to monotherapy darunavir/ritonavir in stable HIV patients: the CINAMMON study. J Neuro-Oncol. 2018;24:98–105. Barber TJ, Imaz A, Boffito M, Niubo J, Pozniak A, Fortuny R, Alonso J, Davies N, Mandalia S, Podzamczer D, Gazzard B. CSF inflammatory markers and neurocognitive function after addition of maraviroc to monotherapy darunavir/ritonavir in stable HIV patients: the CINAMMON study. J Neuro-Oncol. 2018;24:98–105.
62.
go back to reference Parkhurst CN, Yang G, Ninan I, Savas JN, Yates JR 3rd, Lafaille JJ, Hempstead BL, Littman DR, Gan WB. Microglia promote learning-dependent synapse formation through brain-derived neurotrophic factor. Cell. 2013;155:1596–609.CrossRefPubMedCentralPubMed Parkhurst CN, Yang G, Ninan I, Savas JN, Yates JR 3rd, Lafaille JJ, Hempstead BL, Littman DR, Gan WB. Microglia promote learning-dependent synapse formation through brain-derived neurotrophic factor. Cell. 2013;155:1596–609.CrossRefPubMedCentralPubMed
63.
go back to reference Elkabes S, DiCicco-Bloom EM, Black IB. Brain microglia/macrophages express neurotrophins that selectively regulate microglial proliferation and function. J Neurosci. 1996;16:2508–21.CrossRefPubMedCentralPubMed Elkabes S, DiCicco-Bloom EM, Black IB. Brain microglia/macrophages express neurotrophins that selectively regulate microglial proliferation and function. J Neurosci. 1996;16:2508–21.CrossRefPubMedCentralPubMed
64.
go back to reference Bredesen DE, Rabizadeh S. p75NTR and apoptosis: Trk-dependent and Trk-independent effects. Trends Neurosci. 1997;20:287–90.CrossRefPubMed Bredesen DE, Rabizadeh S. p75NTR and apoptosis: Trk-dependent and Trk-independent effects. Trends Neurosci. 1997;20:287–90.CrossRefPubMed
65.
go back to reference Glazner GW, Mattson MP. Differential effects of BDNF, ADNF9, and TNFalpha on levels of NMDA receptor subunits, calcium homeostasis, and neuronal vulnerability to excitotoxicity. Exp Neurol. 2000;161:442–52.CrossRefPubMed Glazner GW, Mattson MP. Differential effects of BDNF, ADNF9, and TNFalpha on levels of NMDA receptor subunits, calcium homeostasis, and neuronal vulnerability to excitotoxicity. Exp Neurol. 2000;161:442–52.CrossRefPubMed
66.
67.
go back to reference Miguez-Burbano MJ, Espinoza L, Bueno D, Vargas M, Trainor AB, Quiros C, Lewis JE, Asthana D. Beyond the brain: the role of brain-derived neurotrophic factor in viroimmune responses to antiretroviral therapy among people living with HIV with and without alcohol use. J Int Assoc Provid AIDS Care. 2014;13:454–60.CrossRefPubMed Miguez-Burbano MJ, Espinoza L, Bueno D, Vargas M, Trainor AB, Quiros C, Lewis JE, Asthana D. Beyond the brain: the role of brain-derived neurotrophic factor in viroimmune responses to antiretroviral therapy among people living with HIV with and without alcohol use. J Int Assoc Provid AIDS Care. 2014;13:454–60.CrossRefPubMed
68.
go back to reference Angelucci F, Ricci V, Pomponi M, Conte G, Mathe AA, Attilio Tonali P, Bria P. Chronic heroin and cocaine abuse is associated with decreased serum concentrations of the nerve growth factor and brain-derived neurotrophic factor. J Psychopharmacol. 2007;21:820–5.CrossRefPubMed Angelucci F, Ricci V, Pomponi M, Conte G, Mathe AA, Attilio Tonali P, Bria P. Chronic heroin and cocaine abuse is associated with decreased serum concentrations of the nerve growth factor and brain-derived neurotrophic factor. J Psychopharmacol. 2007;21:820–5.CrossRefPubMed
69.
go back to reference Bachis A, Campbell LA, Jenkins K, Wenzel E, Mocchetti I. Morphine withdrawal increases brain-derived neurotrophic factor precursor. Neurotox Res. 2017;32:509–17.CrossRefPubMedCentralPubMed Bachis A, Campbell LA, Jenkins K, Wenzel E, Mocchetti I. Morphine withdrawal increases brain-derived neurotrophic factor precursor. Neurotox Res. 2017;32:509–17.CrossRefPubMedCentralPubMed
70.
go back to reference Nosheny RL, Ahmed F, Yakovlev A, Meyer EM, Ren K, Tessarollo L, Mocchetti I. Brain-derived neurotrophic factor prevents the nigrostriatal degeneration induced by human immunodeficiency virus-1 glycoprotein 120 in vivo. Eur J Neurosci. 2007;25:2275–84.CrossRefPubMed Nosheny RL, Ahmed F, Yakovlev A, Meyer EM, Ren K, Tessarollo L, Mocchetti I. Brain-derived neurotrophic factor prevents the nigrostriatal degeneration induced by human immunodeficiency virus-1 glycoprotein 120 in vivo. Eur J Neurosci. 2007;25:2275–84.CrossRefPubMed
71.
go back to reference Bachis A, Major EO, Mocchetti I. Brain-derived neurotrophic factor inhibits human immunodeficiency virus-1/gp120-mediated cerebellar granule cell death by preventing gp120 internalization. J Neurosci. 2003;23:5715–22.CrossRefPubMedCentralPubMed Bachis A, Major EO, Mocchetti I. Brain-derived neurotrophic factor inhibits human immunodeficiency virus-1/gp120-mediated cerebellar granule cell death by preventing gp120 internalization. J Neurosci. 2003;23:5715–22.CrossRefPubMedCentralPubMed
72.
go back to reference Yang K, Perez-Polo JR, Mu XS, Yan HQ, Xue JJ, Iwamoto Y, Liu SJ, Dixon CE, Hayes RL. Increased expression of brain-derived neurotrophic factor but not neurotrophin-3 mRNA in rat brain after cortical impact injury. J Neurosci Res. 1996;44:157–64.CrossRefPubMed Yang K, Perez-Polo JR, Mu XS, Yan HQ, Xue JJ, Iwamoto Y, Liu SJ, Dixon CE, Hayes RL. Increased expression of brain-derived neurotrophic factor but not neurotrophin-3 mRNA in rat brain after cortical impact injury. J Neurosci Res. 1996;44:157–64.CrossRefPubMed
73.
go back to reference Oyesiku NM, Evans CO, Houston S, Darrell RS, Smith JS, Fulop ZL, Dixon CE, Stein DG. Regional changes in the expression of neurotrophic factors and their receptors following acute traumatic brain injury in the adult rat brain. Brain Res. 1999;833:161–72.CrossRefPubMed Oyesiku NM, Evans CO, Houston S, Darrell RS, Smith JS, Fulop ZL, Dixon CE, Stein DG. Regional changes in the expression of neurotrophic factors and their receptors following acute traumatic brain injury in the adult rat brain. Brain Res. 1999;833:161–72.CrossRefPubMed
74.
go back to reference Lebrun-Julien F, Bertrand MJ, De Backer O, Stellwagen D, Morales CR, Di Polo A, Barker PA. ProNGF induces TNFα-dependent death of retinal ganglion cells through a p75NTR non-cell-autonomous signaling pathway. Proc Natl Acad Sci U S A. 2010;107:3817–22.CrossRef Lebrun-Julien F, Bertrand MJ, De Backer O, Stellwagen D, Morales CR, Di Polo A, Barker PA. ProNGF induces TNFα-dependent death of retinal ganglion cells through a p75NTR non-cell-autonomous signaling pathway. Proc Natl Acad Sci U S A. 2010;107:3817–22.CrossRef
75.
go back to reference Kiprianova I, Freiman TM, Desiderato S, Schwab S, Galmbacher R, Gillardon F, Spranger M. Brain-derived neurotrophic factor prevents neuronal death and glial activation after global ischemia in the rat. J Neurosci Res. 1999;56:21–7.CrossRefPubMed Kiprianova I, Freiman TM, Desiderato S, Schwab S, Galmbacher R, Gillardon F, Spranger M. Brain-derived neurotrophic factor prevents neuronal death and glial activation after global ischemia in the rat. J Neurosci Res. 1999;56:21–7.CrossRefPubMed
76.
go back to reference Roux PP, Colicos MA, Barker PA, Kennedy TE. p75 neurotrophin receptor expression is induced in apoptotic neurons after seizure. J Neurosci. 1999;19:6887–96.CrossRefPubMedCentralPubMed Roux PP, Colicos MA, Barker PA, Kennedy TE. p75 neurotrophin receptor expression is induced in apoptotic neurons after seizure. J Neurosci. 1999;19:6887–96.CrossRefPubMedCentralPubMed
77.
go back to reference Dowling P, Ming X, Raval S, Husar W, Casaccia-Bonnefil P, Chao M, Cook S, Blumberg B. Up-regulated p75NTR neurotrophin receptor on glial cells in MS plaques. Neurology. 1999;53:1676–82.CrossRefPubMed Dowling P, Ming X, Raval S, Husar W, Casaccia-Bonnefil P, Chao M, Cook S, Blumberg B. Up-regulated p75NTR neurotrophin receptor on glial cells in MS plaques. Neurology. 1999;53:1676–82.CrossRefPubMed
78.
go back to reference Goutan E, Marti E, Ferrer I. BDNF, and full length and truncated TrkB expression in the hippocampus of the rat following kainic acid excitotoxic damage. Evidence of complex time-dependent and cell-specific responses. Brain Res Mol Brain Res. 1998;59:154–64.CrossRefPubMed Goutan E, Marti E, Ferrer I. BDNF, and full length and truncated TrkB expression in the hippocampus of the rat following kainic acid excitotoxic damage. Evidence of complex time-dependent and cell-specific responses. Brain Res Mol Brain Res. 1998;59:154–64.CrossRefPubMed
79.
go back to reference Mahajan SD, Schwartz SA, Shanahan TC, Chawda RP, Nair MP. Morphine regulates gene expression of alpha- and beta-chemokines and their receptors on astroglial cells via the opioid mu receptor. J Immunol. 2002;169:3589–99.CrossRefPubMed Mahajan SD, Schwartz SA, Shanahan TC, Chawda RP, Nair MP. Morphine regulates gene expression of alpha- and beta-chemokines and their receptors on astroglial cells via the opioid mu receptor. J Immunol. 2002;169:3589–99.CrossRefPubMed
80.
go back to reference Szabo I, Chen XH, Xin L, Adler MW, Howard OM, Oppenheim JJ, Rogers TJ. Heterologous desensitization of opioid receptors by chemokines inhibits chemotaxis and enhances the perception of pain. Proc Natl Acad Sci U S A. 2002;99:10276–81.CrossRefPubMedCentralPubMed Szabo I, Chen XH, Xin L, Adler MW, Howard OM, Oppenheim JJ, Rogers TJ. Heterologous desensitization of opioid receptors by chemokines inhibits chemotaxis and enhances the perception of pain. Proc Natl Acad Sci U S A. 2002;99:10276–81.CrossRefPubMedCentralPubMed
81.
go back to reference 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.CrossRefPubMedCentralPubMed 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.CrossRefPubMedCentralPubMed
82.
go back to reference Chen C, Li J, Bot G, Szabo I, Rogers TJ, Liu-Chen LY. Heterodimerization and cross-desensitization between the μ-opioid receptor and the chemokine CCR5 receptor. Eur J Pharmacol. 2004;483:175–86.CrossRefPubMed Chen C, Li J, Bot G, Szabo I, Rogers TJ, Liu-Chen LY. Heterodimerization and cross-desensitization between the μ-opioid receptor and the chemokine CCR5 receptor. Eur J Pharmacol. 2004;483:175–86.CrossRefPubMed
83.
go back to reference Szabo I, Wetzel MA, Zhang N, Steele AD, Kaminsky DE, Chen C, Liu-Chen LY, Bednar F, Henderson EE, Howard OM, et al. Selective inactivation of CCR5 and decreased infectivity of R5 HIV-1 strains mediated by opioid-induced heterologous desensitization. J Leukoc Biol. 2003;74:1074–82.CrossRefPubMed Szabo I, Wetzel MA, Zhang N, Steele AD, Kaminsky DE, Chen C, Liu-Chen LY, Bednar F, Henderson EE, Howard OM, et al. Selective inactivation of CCR5 and decreased infectivity of R5 HIV-1 strains mediated by opioid-induced heterologous desensitization. J Leukoc Biol. 2003;74:1074–82.CrossRefPubMed
84.
go back to reference Arnatt CK, Falls BA, Yuan Y, Raborg TJ, Masvekar RR, El-Hage N, Selley DE, Nicola AV, Knapp PE, Hauser KF, Zhang Y. Exploration of bivalent ligands targeting putative mu opioid receptor and chemokine receptor CCR5 dimerization. Bioorg Med Chem. 2016;24:5969–87.CrossRefPubMedCentralPubMed Arnatt CK, Falls BA, Yuan Y, Raborg TJ, Masvekar RR, El-Hage N, Selley DE, Nicola AV, Knapp PE, Hauser KF, Zhang Y. Exploration of bivalent ligands targeting putative mu opioid receptor and chemokine receptor CCR5 dimerization. Bioorg Med Chem. 2016;24:5969–87.CrossRefPubMedCentralPubMed
85.
go back to reference Yuan Y, Arnatt CK, El-Hage N, Dever SM, Jacob JC, Selley DE, Hauser KF, Zhang Y. A bivalent ligand targeting the putative mu opioid receptor and chemokine receptor CCR5 heterodimers: binding affinity versus functional activities. Medchemcomm. 2013;4:847–51.CrossRefPubMed Yuan Y, Arnatt CK, El-Hage N, Dever SM, Jacob JC, Selley DE, Hauser KF, Zhang Y. A bivalent ligand targeting the putative mu opioid receptor and chemokine receptor CCR5 heterodimers: binding affinity versus functional activities. Medchemcomm. 2013;4:847–51.CrossRefPubMed
86.
go back to reference El-Hage N, Dever SM, Podhaizer EM, Arnatt CK, Zhang Y, Hauser KF. A novel bivalent HIV-1 entry inhibitor reveals fundamental differences in CCR5-μ-opioid receptor interactions between human astroglia and microglia. AIDS. 2013;27:2181–90.CrossRefPubMed El-Hage N, Dever SM, Podhaizer EM, Arnatt CK, Zhang Y, Hauser KF. A novel bivalent HIV-1 entry inhibitor reveals fundamental differences in CCR5-μ-opioid receptor interactions between human astroglia and microglia. AIDS. 2013;27:2181–90.CrossRefPubMed
Metadata
Title
A central role for glial CCR5 in directing the neuropathological interactions of HIV-1 Tat and opiates
Authors
Sarah Kim
Yun Kyung Hahn
Elizabeth M Podhaizer
Virginia D McLane
Shiping Zou
Kurt F Hauser
Pamela E Knapp
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Journal of Neuroinflammation / Issue 1/2018
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-018-1320-4

Other articles of this Issue 1/2018

Journal of Neuroinflammation 1/2018 Go to the issue