Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Sep 20.
Published in final edited form as: J Neuroimmune Pharmacol. 2011 Aug 18;6(4):528–539. doi: 10.1007/s11481-011-9306-3

Immunomodulatory Properties of Kappa Opioids and Synthetic Cannabinoids in HIV-1 Neuropathogenesis

Shuxian Hu 1,2, Wen S Sheng 3,4, Robert Bryan Rock 5,6
PMCID: PMC8450937  NIHMSID: NIHMS1734306  PMID: 21850403

Abstract

Anti-retroviral therapy (ART) has had a tremendous impact on the clinical outcomes of HIV-1 infected individuals. While ART has produced many tangible benefits, chronic, long-term consequences of HIV infection have grown in importance. HIV-1-associated neurocognitive disorder (HAND) represents a collection of neurological syndromes that have a wide range of functional cognitive impairments. HAND remains a serious threat to AIDS patients, and there currently remains no specific therapy for the neurological manifestations of HIV-1. Based upon work in other models of neuroinflammation, kappa opioid receptors (KOR) and synthetic cannabinoids have emerged as having neuroprotective properties and the ability to dampen pro-inflammatory responses of glial cells; properties that may have a positive influence in HIV-1 neuropathogenesis. The ability of KOR ligands to inhibit HIV-1 production in human microglial cells and CD4 T lymphocytes, demonstrate neuroprotection, and dampen chemokine production in astrocytes provides encouraging data to suggest that KOR ligands may emerge as potential therapeutic agents in HIV neuropathogenesis. Based upon findings that synthetic cannabinoids inhibit HIV-1 expression in human microglia and suppress production of inflammatory mediators such as nitric oxide (NO) in human astrocytes, as well as a substantial literature demonstrating neuroprotective properties of cannabinoids in other systems, synthetic cannabinoids have also emerged as potential therapeutic agents in HIV neuropathogenesis. This review focuses on these two classes of compounds and describes the immunomodulatory and neuroprotective properties attributed to each in the context of HIV neuropathogenesis.

Keywords: HIV-1, Neuropathogenesis, Cannabinoids, Kappa opioid receptors

Background

In 2009, there were an estimated 2.6 million individuals who became newly infected with HIV in the world; this represents a 21% decrease since the peak year of 1997. There were an estimated 1.8 million deaths in 2009, representing a 19% decrease from the peak year of 2004. The success made in reducing acquisition of HIV and improved mortality due to antiretroviral therapy (ART) has translated into an estimated 33.3 million individuals living with HIV (27% increase since 1999) (United Nations Program on HIV/AIDS (UNAIDS)/World Health Organization 2010). With the transformation of HIV from a death sentence to a chronic medical condition for many, long-term consequences of HIV infection have grown in importance. HIV-1-associated neurocognitive disorder (HAND) represents a collection of neurological syndromes that have a wide range of functional cognitive impairments. HAND includes asymptomatic neurocognitive impairment (ANI), mild neurocognitive disorder (MND), and the more severe HIV-associated dementia (HAD). ANI is defined by neuropsychological test results at least one standard deviation below the mean in at least two specific cognitive areas; MND includes meeting criteria for ANI by neuropsychological testing, along with impairment of activities of daily living. The diagnosis of HAD requires neuropsychological test results at least two standard deviations below the mean in two or more cognitive areas and marked impairment of activities of daily living (Gannon et al. 2011).

Despite the therapeutic impact of ART, HAND still represents the most common form of dementia in adults under age 40 and remains an independent risk factor for mortality despite ART (Boisse et al. 2008). As such, it remains a serious threat to AIDS patients (Anthony et al. 2005; Clements et al. 2005; McArthur et al. 2005; Nath and Sacktor 2006; Heaton et al. 2011) and there currently is no specific therapy for the neurological manifestations of HIV-1. Several non-exclusive theories have been proposed as to why ART has not eliminated HAND. They include irreversible brain injury prior to initiating ART, incomplete viral suppression in the CNS due to poor CNS penetration of ART drugs and/or presence of drug-resistant viral strains, possible neurotoxicity of ART drugs, and increased rates of metabolic abnormalities and associated vascular pathology or increased β-amyloid deposition in the brain (Heaton et al. 2011). Another possibility is that even very low levels of viral replication in the CNS could result in neural injury or dysfunction due to prolonged exposure to inflammatory responses and neurotoxic viral proteins (Heaton et al. 2011).

Looking to other models of neuroinflammation for potential compounds that influence HIV-1 neuropathogenesis, synthetic cannabinoids have emerged as having neuroprotective properties and the ability to dampen pro-inflammatory responses of glial cells. Based upon findings that synthetic cannabinoids inhibit HIV-1 expression in human microglia (Peterson et al. 2004; Rock et al. 2007) and suppress production of inflammatory mediators such as nitric oxide (NO) in human astrocytes (Sheng et al. 2005), as well as a substantial literature demonstrating neuroprotective properties of cannabinoids in other systems (Hampson et al. 1998; Shen and Thayer 1998; Nagayama et al. 1999; Chen and Buck 2000; Shohami and Mechoulam 2000; Marsicano et al. 2002, 2003), synthetic cannabinoids have emerged as potential therapeutic agents in HIV neuropathogenesis. Kappa opioid receptor (KOR) ligands have also demonstrated promising anti-inflammatory and neuroprotective properties in several in vitro models of HIV neuropathogenesis. The ability of KOR ligands to inhibit HIV-1 production in human microglial cells (Chao et al. 1996) and CD4 T lymphocytes (Peterson et al. 2001), demonstrate neuroprotection (Chao et al. 2000), and dampen chemokine production in astrocytes (Sheng et al. 2003) provides encouraging data to suggest that KOR ligands may also emerge as potential therapeutic agents in HIV neuropathogenesis.

Research over the past two decades has revealed that a network of factors, including virotoxins gp120 and Tat and mediators released from activated glial cells, are involved in the neuropathogenesis of HIV-1 (D’Aversa et al. 2005; Gendelman 2005; Ghorpade et al. 2005; Gonzalez-Scarano and Martin-Garcia 2005; Kaul et al. 2005; Mattson et al. 2005; Kaul and Lipton 2006). More recent work has demonstrated that the nigrostriatal dopaminergic system is a critical brain region for the neuronal dysfunction and death seen in HAND (Ferris et al.; Aylward et al. 1993; Sardar et al. 1996; Itoh et al. 2000; Nath et al. 2000; Koutsilieri et al. 2002a, b; Wang et al. 2004; Paul et al. 2005; Gelman et al. 2006; Silvers et al. 2006). One interesting feature of the nigrostriatal dopaminergic system (midbrain and the substantia nigra specifically) that may help explain the susceptibility of this brain region to HIV-1-related damage is that this area contains a high concentration of microglial cells (Lawson et al. 1990), the resident macrophages of the brain, which are the only brain cell types that can support productive HIV-1 infection, express CXCR4 and CCR5 and are a rich source of inflammatory mediators (superoxide (O2) and cytokines/chemokines) (Koenig et al. 1986; Wiley et al. 1986; Chao et al. 1995; Cosenza et al. 2002).

Among the cell types in the brain parenchyma, microglia are most capable of generating large quantities of the free radical O2 (Chao et al. 1995), which is rapidly metabolized to the highly cytotoxic reactive oxygen species (ROS) hydrogen peroxide and hydroxyl radical. Unlike microglia from certain rodent species, human microglia do not express inducible nitric oxide synthase (iNOS) (Lee et al. 1993; Peterson et al. 1994; Rock et al. 2005), but when activated, they release abundant amounts of interleukin (IL)-1β, which in turn induces iNOS and NO production by human astrocytes (Hu et al. 1999).

Although not a site of productive viral replication, astrocytes also play an important role in HIV-1 neuropathogenesis (Fine et al. 1996; Conant et al. 1998; Brack-Werner 1999; Persidsky et al. 1999; Lim and Garzino-Demo 2000; Boven et al. 2003; Khurdayan et al. 2004; Kim et al. 2004; Zhou et al. 2004). Astrocytes have been long recognized for their supportive functions in the CNS, including the metabolism of the excitotoxic neurotransmitter glutamate (Danbolt 2001), and we have found that IL-1β-stimulated astrocytes also produce NO (Sheng et al. 2005). Exposure of astrocytes to gp120 impairs the ability of astrocytes to transport L-glutamate (Wang et al. 2003), and glutamate excitotoxicity is thought to be a central end-point pathway in HAD (Lipton and Gendelman 1995). HIV-1 gp120 also creates widespread dysregulation of many other important cellular genes in astrocytes (Su et al. 2002, 2003; Galey et al. 2003).

A growing body of evidence also supports the role of “oxidative stress” in HIV-1 neuropathogenesis (Sheng et al. 2000; Mollace et al. 2001; Aksenov et al. 2003, 2006; Minghetti et al. 2004; Pocernich et al. 2005; Turchan-Cholewo et al. 2006; Wallace et al. 2006). Brain sections obtained from patients with AIDS dementia show intense immunostaining for nitrotyrosine, indicating that reaction between NO and O2 has led to peroxynitrite formation resulting in oxidative damage (Boven et al. 1999). Furthermore, it has been reported that levels of the lipid peroxidation product HNE (4-hydroxy-2-nonenal) are increased in brain tissues of patients with HAD (Haughey et al. 2004).

Compounds that have anti-inflammatory effects on human microglia and astrocytes and demonstrate neuroprotection in the setting of HIV neuropathogenesis may have the potential to supplement ART and specifically address the long-term neurological consequences of HIV infection. In this review, we will address two such classes of compounds: synthetic kappa opioid receptor (KOR) agonists and cannabinoids.

Opioids

The effects of opioids are mediated mainly through opioid receptors classified as mu (μ), kappa (κ), and delta (δ) opioid receptors. A number of opioid agonists and antagonists have been evaluated in in vitro studies and for therapeutic potential in HIV-1 infection. Of these, KORs, which are widely expressed in the central nervous system, have emerged with the most promise (Table 1).

Table 1.

Selected KOR ligands

Class Name
KOR Agonists Butorphanol [17-cyclobutylmethyl-morphinan-3,14-diol]
Spiradoline [2-(3,4-dichlorophenyl)- N-methyl- N-[(5R,7S,8S)- 7-pyrrolidin-1-yl- 1-oxaspiro[4.5]decan-8-yl] acetamide]
Salvinorin A [methyl (2S,4aR,6aR,7R,9S,10aS,10bR)-9-(acetyloxy)-2-(furan-3-yl)-6a,10b-dimethyl-4,10-dioxododecahydro-2H-benzo[f]isochromene-7-carboxylate]
2-methoxymethyl Salvinorin B [(2S,4aR,6aR,7R,9S,10aS,10bR)- 9-(methoxymethoxy)- 2-(3-furanyl) dodecahydro-6a,10b-dimethyl-4,10-dioxo-2H-naphtho-[2,1-c]pyran-7-carboxylic acid methyl ester]
Cyclazocine [2-cyclopropylmethyl- 2’-hydroxy- 5,9-dimethyl- 6,7-benzomorphan]
Menthol [(1R,2S,5R)-2-isopropyl-5-methylcyclohexanol]
Dextromethorphan [(+)-3-methoxy-17-methyl-(9α,13α,14α)-morphinan]
Pentazocine [2-dimethylallyl-5,9-dimethyl-2’-hydroxybenzomorphan]
Norbuprenorphine [(5α,6β,14β,18R)-18-[(1S)-1-hydroxy-1,2,2-trimethylpropyl]-6-methoxy-18,19-dihydro-4,5-epoxy-6,14-ethenomorphinan-3-ol]
Ketazocine [(2S,6R,11R)-3-(cyclopropylmethyl)-8-hydroxy-6,11-dimethyl-3,4,5,6-tetrahydro-2,6-methano-3-benzazocin-1(2H)-one]
Enadoline [2-(benzofuran-4-yl)-N-methyl-N-((5R,7S,8S)-7-(pyrrolidin-1-yl)-1-oxaspiro[4.5] decan-8-yl)ethanamide]
Dynorphin
Asimadoline [N-[(1S)-2-[(3S)-3-hydroxypyrrolidin-1-yl]-1-phenylethyl]-N-methyl-2,2-diphenylacetamide]
Tifluadom [N-[(5-(2-fluorophenyl)- 1-methyl- 2,3-dihydro- 1,4-benzodiazepin-2-yl) methyl] thiophene- 3-carboxamide]
Nalfurafine [(2E)-N-[(5α,6β)-17-(cyclopropylmethyl)- 3,14-dihydroxy- 4,5-epoxymorphinan- 6-yl]- 3-(3-furyl)-N-methylacrylamide]
HZ-2 [Dimethyl 3,7-dimethyl-9-oxo-2,4-dipyridin-2-yl-3,7-diazabicyclo[3.3.1]nonane-1,5-dicarboxylate]
Nalbuphine [(−)-17-(cyclobutylmethyl)- 4,5α-epoxymorphinan- 3,6α,14-triol hydrochloride]
LPK-26 [2-(3,4-dichlorophenyl)-N-[(2S)-1-(2,5-dihydropyrrol-1-yl)-3-methylbutan-2-yl]-N-methylacetamide]
BRL-52537 [2-(3,4-dichlorophenyl)-1-[(2S)-2-(pyrrolidin-1-ylmethyl)piperidin-1-yl]ethanone]
ICI-204,448 [2-(3-[1-([2-(3,4-dichlorophenyl)acetyl]-methylamino)-2-pyrrolidin-1-ylethyl]phenoxy)acetic acid]
ICI-199,441 [2-(3,4-dichlorophenyl)-N-methyl-N-[(1S)-1-phenyl-2-pyrrolidin-1-ylethyl]acetamide]
U50,488 [trans-3,4-dichloro-N-methyl-N[2-(1-pyrolidinyl)cyclohexyl]benze neaceamide methanesulfonate]a
U69,593 [N-methyl-2-phenyl-N-[(5R,7S,8S)-7-(pyrrolidin-1-yl)-1-oxaspiro[4.5]dec-8-yl]acetamide]a
KOR Antagonists 5’ Guanidinonaltrindole [5’-Guanidinyl-17-(cyclopropylmethyl)-6,7-dehydro-4,5α-epoxy-3,14-dihydroxy-6,7–2’,3’-indolomorphinan]
Buprenorphine [(2S)-2-[(−)-(5R,6R,7R,14S)-9α-cyclopropylmethyl-4,5-epoxy-6,14-ethano-3-hydroxy-6-methoxymorphinan-7-yl]-3,3-dimethylbutan-2-ol]
nor-BNI [Norbinaltorphimine] [17,17’-(dicyclopropylmethyl)-6,6’,7,7’−6,6’-imino- 7,7’-binorphinan-3,4’,14,14’-tetrol]
JDTic [(3R)-7-hydroxy-N-((1S)-1-{[(3R,4R)-4- (3-hydroxyphenyl)- 3,4-dimethylpiperidin- 1-yl]methyl}- 2-methylpropyl)- 1,2,3,4-tetrahydroisoquinoline- 3-carboxamide]

A list of KOR agonists and antagonists

a

Activity demonstrated in HIV neuropathogenesis as described in the text

There are several lines of evidence to suggest that KOR ligands may have therapeutic effects on HIV neuropathogenesis. Many years ago, our laboratory reported the presence of KORs in human fetal microglia and inhibition of HIV-1 expression in acutely infected microglial cell cultures treated with the KOR ligands U50,488 and U69,593. This antiviral effect of kappa ligands was blocked by the specific KOR antagonist, nor-binaltrophimine (nor-BNI) (Chao et al. 1996). We later investigated whether selective KOR ligands would exert antiviral effects in acutely infected mixed glial/neuronal cell cultures. The proinflammatory cytokines TNF-α and IL-1β have been found to profoundly suppress HIV-1 expression in this mixed glial/neuronal brain cell culture model (Lokensgard et al. 1997). While the KOR ligand U50,488 alone had little anti-HIV-1 activity, this opioid potentiated in a concentration-dependent manner the antiviral activity of tumor necrosis factor (TNF)-α, but not the antiviral activity of IL-1β. The KOR antagonist nor-BNI completely blocked the potentiating effect of U50,488 on TNF-α, suggesting the involvement of a KOR-mediated mechanism. Antibodies to TNF-α completely blocked the potentiating effect of U50,488, suggesting that the potentiating effect of U50,488 is dependent upon TNF-α. While U50,488 did not potentiate the antiviral activity of IL-1β, antibodies to IL-1β blocked the potentiating effect of U50,488 on the antiviral activity of TNF-α, suggesting that IL-1β also contributes to the potentiating effect of U50,488 on TNF-α (Chao et al. 1998). Again using primary human brain cell cultures, we found that U50,488 also suppressed in a dose-dependent manner the neurotoxicity mediated by supernatants derived from HIV-1-infected microglia. This neuroprotective effect of U50,488 was blocked by the KOR antagonist nor-BNI. The neurotoxic activity of the supernatants from HIV-1-infected microglia was also blocked by the NMDA receptor antagonists 2-amino-5-phosphonovalerate (2-APV) and MK-801. HIV-1 infection of microglial cell cultures induced the release of quinolinate, and U50,488 dose-dependently suppressed quinolinate release by infected microglial cell cultures with a corresponding inhibition of HIV-1 p24 antigen levels (Chao et al. 2000). Treatment of astrocytes with U50,488 inhibited Tat-induced CCL-2 production in a concentration-dependent manner. The KOR antagonist nor-BNI completely blocked the inhibitory effect of U50,488, suggesting involvement of KOR. While U50,488 alone had a partial inhibitory effect on constituent NF-κB activation, it potently suppressed Tat-induced NF-κB activation (Sheng et al. 2003). Overall, these in vitro findings suggest that KOR agonists have immunomodulatory activity in the brain and may have therapeutic potential in HIV-1 neuropathogenesis by attenuating microglial cell production of the neurotoxin quinolinate and viral proteins and dampening chemokine production in astrocytes.

Because infiltrating monocytes and T-cells contribute to HIV neuropathogenesis, we and others also investigated the immunomodulatory effects of KOR ligands in these cell populations. In one study, peritoneal macrophages were treated simultaneously with U50,488 and lipopolysaccharide (LPS), and the levels of the cytokines IL-1, IL-6 and TNF-α were measured. The results showed that U50,488 had a suppressive effect on the production of TNF-α, IL-1, and IL-6. Naloxone was able to partially block U50,488 suppression, while nor-BNI was able to completely reverse the suppression of IL-6 production (Alicea et al. 1996). Our laboratory investigated whether U50,488 would exert such an anti-HIV-1 effect in acutely infected blood monocyte-derived macrophages (MDM). Treatment of HIV-1-infected MDM with U50,488 induced a concentration-dependent inhibition of HIV-1 expression. The dose—response relationship of U50,488 was U-shaped with a peak effect observed at 10−13 M. The KOR antagonist nor-BNI blocked this anti-HIV-1 effect, indicating involvement of KORs; further studies suggested that the anti-HIV-1 effect of U50,488 partially involved the production of CCL5 by MDM (Chao et al. 2001).

CD4 lymphocytes are a primary cell target for HIV-1, and these cells are known to express opioid receptors. In our laboratory, activated CD4 lymphocytes were infected with HIV-1, and p24 antigen levels were measured in supernatants of naltrexone-treated or untreated cultures. While naltrexone alone did not affect HIV-1 expression in activated CD4 lymphocytes, naltrexone increased the antiviral activity of AZT and indinavir by 2–3-fold. Similar findings with a KOR selective antagonist supported the possible involvement of KOR in naltrexone’s potentiation of the antiretroviral drugs (Gekker et al. 2001). Beyond the effect on antiretroviral drugs, we also examined the effects of U50,488 on HIV-1 expression in acutely infected CD4 lymphocytes. When U50,488 was added to activated CD4+ lymphocytes, HIV-1 expression was inhibited in a concentration- and time-dependent manner. The KOR antagonist nor-BNI had no effect by itself on viral expression but blocked the antiviral property of U50,488, suggesting that U50,488 was acting via a KOR (Peterson et al. 2001).

The last study above showed that the suppressive effect of KOR ligands on HIV-1 expression in acutely infected CD4 lymphocytes is concentration and time-dependent. This finding implies that the inhibition by U50,488 occurs at an early step in the viral replication cycle. In a follow up study, we hypothesized that U50,488 treatment of CD4 lymphocytes inhibited HIV-1 envelope (Env) glycoprotein-mediated membrane fusion. To address this we used a vaccinia virus-based assay to measure the effects of U50,488 treatment of CD4 lymphocytes on HIV-1IIIB Env glycoprotein-mediated fusogenic activity, based on the cytoplasmic activation of a reporter gene. The results showed that U50,488 inhibited Env-mediated cell fusion in a bell-shaped concentration-response, which suggests that the effect operates in a very narrow therapeutic range and is likely quite complex. The KOR antagonist nor-BNI blocked of the inhibitory activity of U50,488, implying that U50,488 was acting via a KOR-related mechanism. Using flow cytometry, we demonstrated that the chemokine co-receptor CXCR4, but not CD4, was down-regulated as a consequence of KOR activation. These findings support the hypothesis that KOR activation on CD4 lymphocytes inhibits HIV-1 entry via down-regulation of CXCR4 (Lokensgard et al. 2002).

We also examined the effects of KOR ligands on the deleterious effects of other drugs of abuse on HIV neuropathogenesis. Treatment of microglia with cocaine promoted HIV-1 expression and pretreatment of microglia with KOR agonists as well as nor-BNI abrogated the cocaine-induced potentiation of viral expression. The mechanism whereby KOR ligands inhibit cocaine’s stimulatory effect on viral expression involves the suppression of cocaine-induced activation of extracellular signal-regulated kinase1/2, thereby blunting cocaine-enhanced up-regulation of the HIV-1 entry chemokine co-receptor CCR5 (Gekker et al. 2004).

More recently, we looked at the effects of KOR ligands on neural precursor cells (NPCs). NPCs are self-renewing, multipotent cells capable of differentiating into neurons, astrocytes, and oligodendrocytes and have been shown to migrate toward sites of injury in the brain and to participate in the process of brain repair. We documented robust expression of KORs in highly enriched (>90% nestin-positive) human fetal brain-derived NPCs. We also found that U50,488, but not dynorphin(2–17), stimulated proliferation and migration of NPCs in a concentration-dependent manner. The KOR antagonist, nor-BNI, partially blocked the migratory and proliferative effects of KOR agonists supporting the involvement of a KOR-related mechanism (Sheng et al. 2007).

One of the proposed mechanisms by which KOR ligands may induce their immunomodulatory effects on HIV involves the ability of KOR to cross-desensitize the HIV-1 co-receptor CXCR4 in a bi-directional fashion (Finley et al. 2008). In a recent study using a combination of biochemical approaches, these investigators showed that both CXCR4 protein and mRNA levels are significantly reduced following KOR activation. They also determined that the IRFs (interferon regulated factors) and STATs (signal transducer and activator of transcription) were induced following KOR activation, that JAK2 (Janus Kinase), STAT3, and IRF2 were critical members of this signal transduction pathway and that these processes were important for the inhibition of CXCR4 expression (Finley et al. 2011).

Cannabinoids

Of the two classical cannabinoid receptors, cannabinoid receptor 1 (CB1) receptors are expressed predominantly in the brain, and are abundantly expressed in the basal ganglia and hypothalamus. Activation of these receptors by natural cannabinoids, such as Δ9-tetrahydrocannabinol (THC), as well as by synthetic cannabinoids has been extensively investigated (Howlett et al. 2004). Cannabinoid receptor 2 (CB2) receptors are predominantly expressed in cells of the immune system, including microglia (Klein and Cabral 2006; Raborn and Cabral 2010) (Table 2).

Table 2.

Selected cannabinoid ligands

Class Name CB1 activity CB2 activity
Phytocannabinoids THC [Δ9-tetrahydrocannabinol] [(−)-(6aR,10aR)-6,6,9-trimethyl-3-pentyl-6a,7, 8,10a-tetrahydro-6H-benzo[c]chromen-1-ol]a Agonist Agonist
CBD [Cannabidiol] [2-[(1R,6R)-6-isopropenyl-3-methylcyclohex-2-en-1-yl]-5-pentylbenzene-1,3-diol] Indirect Antagonist Indirect Antagonist
Endogenous Cannabinoids 2-AG [2-Arachidonoylglycerol] [1,3-Dihydroxy-2-propanyl (5Z,8Z,11Z,14Z)-5,8,11,14-eicosatetraenoate] Agonist Agonist
Anandamide [N-arachidonoylethanolamine or AEA] [(5Z,8Z,11Z,14Z)-N-(2-hydroxyethyl)icosa-5,8,11,14-tetraenamide]a Agonist Agonist
Synthetic Cannabinoids (+)-WIN 55,212-2 [(4,5-dihydro-2-methyl-4(4-morpholinylmethyl)-1-(1-naphthalenyl-carbonyl)- 6H-pyrrolo[3,2,1ij]quinolin-6-one]a Agonist Agonist
CP55,940 [(−)-cis-3-[2-hydroxy-4-(1,1-dimethylheptyl)phenyl]-trans-4-(3-hydroxypropy l) cyclohexanol]a Agonist Agonist
HU-210 [(6aR,10aR)- 9-(Hydroxymethyl)- 6,6-dimethyl- 3-(2-methyloctan-2-yl)-6a, 7,10,10a-tetrahydrobenzo [c]chromen- 1-ol] Agonist Agonist
JWH-018 [(1-pentyl-3-(1-naphthoyl)indole) or AM-67] Agonist Agonist
ACEA [N-(2-chloroethyl)-5Z,8Z,11Z,14Z-eicosatetraenamide]a Agonist
O-2137 [(1R,3R)-1-[4-(1,1-dimethylheptyl)-2,6-dimethoxyphenyl]-3 methylcyclohexanol] Agonist
JW-133 [(6aR,10aR)-3-(1,1-Dimethylbutyl)-6a,7,10,10a-tetrahydro −6,6,9-trimethyl-6H-dibenzo[b,d]pyran] Agonist
JWH-015 [(2-Methyl-1-propyl-1H-indol-3-yl)-1-naphthalenylmethanone]a Agonist
SR141716A [5-(4-chlorophenyl)-1-(2,4-dichlorophenyl)-4-methyl-N-(1-piperidyl)pyrazole −3-carboxamide hydrochloride] Antagonist
SR144528 [(1S-endo)-5-(4-chloro-3-methylphenyl)-1-((4-methylphenyl)methyl)-N-(1,3,3- trimethylbicyclo(2.2.1)hept-2-yl)-1H-pyrazole-3-carboxamide] Antagonist

A list of phytocannabinoids, endogenous cannabinoids, and synthetic cannabinoids with references to the dominant receptor binding activity

a

Activity demonstrated in HIV neuropathogenesis as described in the text

The protective effects of cannabinoids on neuronal cell damage have been extensively studied in several rodent models (Facchinetti et al. 2003; Klegeris et al. 2003; Walter et al. 2003; Eljaschewitsch et al. 2006; Klein and Cabral 2006). These studies demonstrate that both CB1 receptor-mediated (Shen and Thayer 1998; Nagayama et al. 1999; Marsicano et al. 2003) and non-receptor-mediated (antioxidant effects) (Chen and Buck 2000; Shohami and Mechoulam 2000; Marsicano et al. 2002) mechanisms are involved in the neuroprotective properties of cannabinoids. Cannabinoids alter immune cell functions (Klein et al. 2003), including certain functions of microglia, the resident macrophages of the brain parenchyma (Cabral and Griffin-Thomas 2008). These activities appear to be mediated through cannabinoid receptors (CB1 or CB2). Synthetic cannabinoid agonists have a beneficial effect in animal models of several neurodegenerative diseases, including multiple sclerosis (Croxford and Miller 2003), Parkinson’s disease (Price et al. 2009), and Huntington’s disease (Palazuelos et al. 2009).

Several studies have demonstrated the potential importance of cannabinoid receptors in HIV neuropathogenesis. A recent in vivo experiment linked the thermoregulatory effects of the synthetic cannabinoid, WIN55,212–2, which produced a dose-related hypothermia, and the HIV-1 co-receptor CXCR4 (Benamar et al. 2009). Using quantitative image analysis and immunohistochemistry in the cerebral cortex and white matter of patients with HIV encephalitis (HIVE), the authors observed that CB1 was increased in HIVE brains, while CB2 was significantly increased in the white matter of HIVE. At baseline, CB1 was noted in neurons, and both CB1 and CB2 were noted in meningeal macrophages and subpial glia. In HIVE, CB1 was found in white matter microglia and perivascular cells, while CB2 was increased in perivascular macrophages, microglia, and astrocytes (Cosenza-Nashat et al. 2011).

The current use of cannabinoids in clinical medicine has focused on the treatment of neuropathic pain, nausea and anorexia. Dronabinol (Marinol), a synthetic THC, is used as an appetite stimulant, anti-emetic, and analgesic. It also has been used for HIV-wasting disease since the early years of the HIV epidemic (Timpone et al. 1997). Sativex, a cannabinoid extract oral spray containing THC, cannabidiol (CBD), and other cannabinoids is used for the treatment of neuropathic pain and spasticity in 22 countries including England, Canada and Spain. Nabilone (Cesamet), a synthetic cannabinoid is used as an antiemetic and for the treatment of neuropathic pain. Rimonabant (SR141716A), a selective CB1 receptor antagonist, was used as an anti-obesity drug under the proprietary name Acomplia in the European Union until the use of this product was suspended in 2009.

One of the initial concerns regarding cannabinoid use among HIV-infected individuals, whether for recreational or therapeutic purposes, has centered on whether exogenous cannabinoids would worsen HIV-1 infection itself. One group investigated whether cannabinoids enhanced or suppressed HIV-1 infection in several cell lines (SupT, and H9, H9MN, and MT-2 cells). Of the cell lines tested, only the MT-2 cells were positive for both CB1 and CB2 mRNA. Using CP55,940, THC, WIN55,212–2, and WIN55,212–3 as their candidate cannabinoids, the authors attempted to determine whether different cannabinoid receptor agonists influenced infection of these cells by cell free HIV-1. Using syncytia formation as an indication of virus infection and cytopathicity, the authors concluded that cannabinoids may enhance HIV-1 infection of susceptible cells (Noe et al. 1998). In another study using a short-term exposure model of human endothelium, monocyte adherence was diminished with morphine and anandamide treatment, whereas monocyte adherence was enhanced with gp120. Short-term exposure to morphine or anandamide also resulted in nitric oxide (NO) release, whereas exposure to gp120 did not. Long-term exposure of endothelial cells to gp120, morphine or anandamide resulted in enhancement of monocyte adherence; these effects were further increased when exposed to gp120 in combination with either morphine or anandamide. The authors concluded that this was caused by a desensitization of the endothelium to further NO release after the initial exposure to either morphine or anandamide. From these results the authors concluded that exposure to morphine or cannabinoids may cause HIV to progress more rapidly because monocyte adherence and mobility is significantly increased, indicating a higher level of trans-membrane migration (Stefano et al. 1998a).

Based on further observations of macrophage behavior during chronic exposure to morphine and anandamide, these agents first induced a period of inactivity which was followed by a period of chemokinesis; NO was released by macrophages only when exposed to morphine and anandamide, but not gp120. Thus, the authors concluded that the chemokinetic inducing activities of morphine and anandamide may be the basis for excitotoxin liberation in neural tissues and/or a higher HIV-1 viral load (Stefano et al. 1998b). In an in vitro study investigating the effects of THC and CBD on cytokine production by human leukemic T, B, eosinophilic and CD8+ NK cell lines as models they found that THC decreased constitutive production of CXCL8, CCL3, CCL4, and CCL5 and phorbol ester stimulated production of TNF-α, granulocyte macrophage colony stimulating factor (GM-CSF) and IFN-γ by NK cells. THC also inhibited CCL4 in human T-cell lymphotrophic virus type 1 (HTLV-1) positive B-cells but increased CXCL8, CCL3 and CCL4 in B-cells and CCL4 and CXCL8 in eosinophils. Both cannabinoids strongly inhibited IL-10 production by HUT-78 T-cells. The authors cautioned that because of the mixed and complex effects that these two cannabinoids have on cytokine production by various cell lines, cannabinoids may benefit some diseases while worsening others (Srivastava et al. 1998). Finally, in a study using a hu-PBL-SCID mouse model (human peripheral blood leukocytes [PBL] implanted into severe combined immunodeficient mice[SCID]) and infected with an HIV reporter construct in the presence or absence of THC exposure, it was suggested that exposure to THC in vivo can suppress immune function, increase HIV co-receptor expression, and act as a cofactor to significantly enhance HIV replication (Roth et al. 2005).

The concerns regarding whether exogenous cannabinoids would worsen HIV-1 infection have been tempered considerably with more recent data. Based on clinical data, there does not appear to be any significant effect of cannabinoid use among HIV-infected individuals regarding HIV RNA, CD4/CD8 levels and immune phenotypes, at least in the short term (Bredt et al. 2002; Abrams et al. 2003). In a randomized placebo-controlled study designed to evaluate the metabolic effects of smoked marijuana and dronabinol in HIV-infected patients receiving two early-era antiretroviral drugs, indinavir and nelfinavir, the authors concluded that despite a decrease in the maximum concentration of indinavir in those in the marijuana arm of the study, the magnitude of the observed pharmacokinetic changes was unlikely to impact antiretroviral efficacy of these two agents (Kosel et al. 2002).

Results of several in vivo models of HIV suggest cannabinoids may actually have beneficial effects. Chronic THC administration decreased early mortality from simian immunodeficiency virus (SIV) infection, and this was associated with attenuation of plasma and CSF viral load and retention of body mass (Molina et al. 2010). In vitro, THC decreased SIV viral replication in MT4-R5 cells (Molina et al. 2010). Chronic use of THC (12 months) did not alter lymphocyte subtypes, naive and memory subsets, proliferation, or apoptosis of T lymphocytes in male young adult rhesus macaques. However, chronic THC increased T lymphocyte CXCR4 expression on both CD4+ and CD8+ T lymphocytes compared to controls (Lecapitaine et al. 2011). In a feline immunodeficiency virus (FIV) model, macrophages were shown to express cannabinoid receptors, and recombinant feline interferon-γ and lipopolysaccharide (LPS)-induced NO production decreased after in vitro exposure to the synthetic cannabinoid CP55,940. This observation involved both CB1 and CB2 receptors (Ponti et al. 2001). In a model where hu-PBL were reconstituted into immunodeficient Balb/c-Rag−/−γc−/− mice and HIV-1-infected human MDMs were injected into the brain to induce viral encephalitis (hu-PBL/HIVE mice), the authors report that the hu-PBL/HIVE mice exhibit altered CB2 receptor expression; no changes in CB1 receptor and GPR55 expression were observed. Gp1a diminished microglial activation and decreased proinflammatory cytokine TNF-α expression in HIVE brains. The significant reduction in GVH-mediated deaths with Gp1a treatment suggested an immune-suppressive effect of Gp1a. There was a decrease in CCR5 expression on CD4+ cells with HIV infection in hu-PBL/HIVE mice compared to hu-PBL mice, which was thought due to the progressive depletion of activated CD4+ cells with infection. Overall the use of Gp1a did not reduce peripheral viral load in this model. Decreased expression of HIV-gag was observed in the brains of Gp1a-treated animals, but was not significant (Gorantla et al. 2010).

Using a model of the human blood brain barrier involving co-cultures of human brain microvascular endothelial cells (HBMEC) and human astrocytes, the authors showed that the cannabinoid agonists CP55,940 and ACEA: 1) inhibited HIV-1 gp120-induced calcium influx mediated by substance P, 2) decreased the permeability of HBMEC, 3) prevented tight junction protein down-regulation and 4) inhibited the transmigration of human monocytes across the BBB and blocked the BBB permeability in vivo. These results demonstrate that synthetic cannabinoids are capable of restoring the integrity of HBMEC following HIV-1 gp120-induced toxicity (Lu et al. 2008).

In our laboratory, we have found that suppression of HIV-1 expression in human microglial cells by the synthetic CB1/CB2 receptor agonists WIN55,212–2 and CP55,940 involves CB2 receptors (Peterson et al. 2004; Rock et al. 2007). Evidence for this conclusion was provided by the findings that the CB2 receptor selective agonist JWH-015 inhibited HIV-1 expression and that the CB2 receptor selective antagonist SR144528 blocked WIN55,212–2’s inhibitory effect. Surprisingly, the CB1 receptor selective antagonist SR141716A behaved as an agonist in this culture system, suggesting human microglia possess functionally active non-CB1/CB2 receptors (Rock et al. 2007).

The inhibitory effect of WIN55,212-2 on IL-1β-stimulated expression of inducible nitric oxide synthase (iNOS) and cytokines/chemokines (TNF-α, CXCL10, CCL2, and CCL5) by human astrocytes, on the other hand was shown to be partially blocked by SR141716A and SR144528 suggesting that these glial cells possess functionally active CB1 and CB2 receptors (Sheng et al. 2005). In an additional study, we found that production of CX3CL1 by human astrocytes stimulated with IL-1β was inhibited in a concentration-dependent manner following pretreatment with the synthetic cannabinoid WIN55,212-2. The CB2 receptor selective antagonist SR144528 significantly inhibited WIN55,212-2-mediated suppression of CX3CL1, suggesting a CB2-receptor-related mechanism. IL-1β triggered the activation of p38 and ERK1/2 (p44/42) MAP kinase (MAPK) signaling pathways, but WIN55,212-2 mainly inhibited p38 MAPK phosphorylation. This finding was mirrored in experiments using known inhibitors of these MAPKs, suggesting that the suppression of CX3CL1 production by WIN55,212-2 involves inhibition of signaling via p38 MAPK (Sheng et al. 2009).

Several mechanisms by which cannabinoids may exert a beneficial effect on HIV neuropathogenesis have been suggested. Using both pharmacological and biochemical knockdown methods, Raborn et al. demonstrated that the cannabinoids THC and CP55,940 inhibited migration of human U937 macrophage-like cells to the HIV-1 Tat protein, and that this modulation of macrophage migration was CB2 mediated (Raborn and Cabral 2010). The CB2 agonist JWH-015 suppressed IFN-γ-induced CD40 expression and phosphorylation of JAK/STAT1in microglial cells, inhibited microglial cell TNF-α and NO production induced either by IFN-γ or Aβ peptide, and attenuated CD40-mediated inhibition of microglial phagocytosis of Aβ1–42 peptide (Ehrhart et al. 2005). WIN 55,212–2 stimulation of Tat + IFN-γ-induced expression of iNOS, NO release and NO-mediated cell toxicity in rat glioma C6 cells. HIV-1 Tat + IFN-γ also induced a significant inhibition of CB1, but not CB2, receptor expression and inhibited anandamide uptake by C6 cells with no effect on anandamide hydrolysis. The authors concluded that stimulation of the endocannabinoid system can reduce HIV-1 Tat-induced cytotoxicity, and is itself regulated by HIV-1 Tat (Esposito et al. 2002).

Final thoughts

HAND remains a significant threat to HIV-1-infected patients, and specific therapy of this crippling neurodegenerative disorder is lacking. This review focused on two potential classes of compounds, synthetic KOR ligands and synthetic cannabinoids, which may be exploited for their neuroprotective properties and abilities to dampen pro-inflammatory responses of glial cells. While early work on these compounds is encouraging, there remain many gaps in knowledge that need to be filled. For KOR ligands, while the in vitro findings are promising, evidence for direct in vivo neuroprotective effects are still lacking, both in terms of confirmation of the in vitro effects, and demonstration of a meaningful, positive effect on disease progression. This lack of in vivo work limits our understanding of the clinical viability of this class of compounds. The work thus far on the neuroprotective role of synthetic cannabinoids has focused on mechanisms related to the CB1 and CB2 receptors, non-CB1/CB2-receptor mediated mechanisms must be considered as well, especially since candidate CB receptors are emerging (Cabral and Griffin-Thomas 2009). While short-term clinical studies have not demonstrated a deleterious effect of cannabinoids on HIV disease (Bredt et al. 2002; Kosel et al. 2002; Abrams et al. 2003), and much of the evidence supporting the neuroprotective effects of synthetic cannabinoids is also based on in vitro studies, encouraging evidence has emerged from studies using and SIV monkey models and hu-PBL/HIVE mouse models (Gorantla et al. 2010; Molina et al. 2010). Additional in vivo investigations would serve to flesh out the potential role of synthetic compounds in a treatment modality targeting HIV neuropathogenesis. While significant effort has been made to study and examine the role of these compounds in HIV neuropathogenesis, much work remains to fully elucidate their potential.

Acknowledgments

We would like to thank Phillip K. Peterson for his significant scientific insight and counsel.

This work was supported, in part by U.S. Public Health Service grant DA025525

Footnotes

None of the authors has a commercial or other association that might pose a conflict of interest with the current review.

Contributor Information

Shuxian Hu, Center for Infectious Diseases and Microbiology Translational Research, University of Minnesota Medical School, Minneapolis, MN, USA; Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, USA.

Wen S. Sheng, Center for Infectious Diseases and Microbiology Translational Research, University of Minnesota Medical School, Minneapolis, MN, USA Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, USA.

Robert Bryan Rock, Center for Infectious Diseases and Microbiology Translational Research, University of Minnesota Medical School, Minneapolis, MN, USA; Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, USA.

References

  1. Abrams DI, Hilton JF, Leiser RJ, Shade SB, Elbeik TA, Aweeka FT, Benowitz NL, Bredt BM, Kosel B, Aberg JA, Deeks SG, Mitchell TF, Mulligan K, Bacchetti P, McCune JM, Schambelan M (2003) Short-term effects of cannabinoids in patients with HIV-1 infection: a randomized, placebo-controlled clinical trial. Ann Intern Med 139:258–266 [DOI] [PubMed] [Google Scholar]
  2. Aksenov MY, Hasselrot U, Wu G, Nath A, Anderson C, Mactutus CF, Booze RM (2003) Temporal relationships between HIV-1 Tat-induced neuronal degeneration, OX-42 immunoreactivity, reactive astrocytosis, and protein oxidation in the rat striatum. Brain Res 987:1–9 [DOI] [PubMed] [Google Scholar]
  3. Aksenov MY, Aksenova MV, Nath A, Ray PD, Mactutus CF, Booze RM (2006) Cocaine-mediated enhancement of Tat toxicity in rat hippocampal cell cultures: the role of oxidative stress and D1 dopamine receptor. Neurotoxicology 27:217–228 [DOI] [PubMed] [Google Scholar]
  4. Alicea C, Belkowski S, Eisenstein TK, Adler MW, Rogers TJ (1996) Inhibition of primary murine macrophage cytokine production in vitro following treatment with the kappa-opioid agonist U50,488H. J Neuroimmunol 64:83–90 [DOI] [PubMed] [Google Scholar]
  5. Anthony IC, Ramage SN, Carnie FW, Simmonds P, Bell JE (2005) Influence of HAART on HIV-related CNS disease and neuroinflammation. J Neuropathol Exp Neurol 64:529–536 [DOI] [PubMed] [Google Scholar]
  6. Aylward EH, Henderer JD, McArthur JC, Brettschneider PD, Harris GJ, Barta PE, Pearlson GD (1993) Reduced basal ganglia volume in HIV-1-associated dementia: results from quantitative neuro-imaging. Neurology 43:2099–2104 [DOI] [PubMed] [Google Scholar]
  7. Benamar K, Yondorf M, Geller EB, Eisenstein TK, Adler MW (2009) Physiological evidence for interaction between the HIV-1 co-receptor CXCR4 and the cannabinoid system in the brain. Br J Pharmacol 157:1225–1231 [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Boisse L, Gill MJ, Power C (2008) HIV infection of the central nervous system: clinical features and neuropathogenesis. Neurol Clin 26:799–819, x [DOI] [PubMed] [Google Scholar]
  9. Boven LA, Gomes L, Hery C, Gray F, Verhoef J, Portegies P, Tardieu M, Nottet HS (1999) Increased peroxynitrite activity in AIDS dementia complex: implications for the neuropathogenesis of HIV-1 infection. J Immunol 162:4319–4327 [PubMed] [Google Scholar]
  10. Boven LA, Vergnolle N, Henry SD, Silva C, Imai Y, Holden J, Warren K, Hollenberg MD, Power C (2003) Up-regulation of proteinase-activated receptor 1 expression in astrocytes during HIV encephalitis. J Immunol 170:2638–2646 [DOI] [PubMed] [Google Scholar]
  11. Brack-Werner R (1999) Astrocytes: HIV cellular reservoirs and important participants in neuropathogenesis. AIDS 13:1–22 [DOI] [PubMed] [Google Scholar]
  12. Bredt BM, Higuera-Alhino D, Shade SB, Hebert SJ, McCune JM, Abrams DI (2002) Short-term effects of cannabinoids on immune phenotype and function in HIV-1-infected patients. J Clin Pharmacol 42:82S–89S [DOI] [PubMed] [Google Scholar]
  13. Cabral GA, Griffin-Thomas L (2008) Cannabinoids as therapeutic agents for ablating neuroinflammatory disease. Endocr Metab Immune Disord Drug Targets 8:159–172 [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Cabral GA, Griffin-Thomas L (2009) Emerging role of the cannabinoid receptor CB2 in immune regulation: therapeutic prospects for neuroinflammation. Expert Rev Mol Med 11:e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Chao CC, Hu S, Peterson PK (1995) Modulation of human microglial cell superoxide production by cytokines. J Leukoc Biol 58:65–70 [DOI] [PubMed] [Google Scholar]
  16. Chao CC, Gekker G, Hu S, Sheng WS, Shark KB, Bu DF, Archer S, Bidlack JM, Peterson PK (1996) kappa opioid receptors in human microglia downregulate human immunodeficiency virus 1 expression. Proc Natl Acad Sci U S A 93:8051–8056 [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Chao CC, Gekker G, Hu S, Kravitz F, Peterson PK (1998) Kappa-opioid potentiation of tumor necrosis factor-alpha-induced anti-HIV-1 activity in acutely infected human brain cell cultures. Biochem Pharmacol 56:397–404 [DOI] [PubMed] [Google Scholar]
  18. Chao CC, Hu S, Gekker G, Lokensgard JR, Heyes MP, Peterson PK (2000) U50,488 protection against HIV-1-related neurotoxicity: involvement of quinolinic acid suppression. Neuropharmacology 39:150–160 [DOI] [PubMed] [Google Scholar]
  19. Chao CC, Gekker G, Sheng WS, Hu S, Peterson PK (2001) U50488 inhibits HIV-1 expression in acutely infected monocyte-derived macrophages. Drug Alcohol Depend 62:149–154 [DOI] [PubMed] [Google Scholar]
  20. Chen Y, Buck J (2000) Cannabinoids protect cells from oxidative cell death: a receptor-independent mechanism. J Pharmacol Exp Ther 293:807–812 [PubMed] [Google Scholar]
  21. Clements JE, Li M, Gama L, Bullock B, Carruth LM, Mankowski JL, Zink MC (2005) The central nervous system is a viral reservoir in simian immunodeficiency virus-infected macaques on combined antiretroviral therapy: a model for human immunodeficiency virus patients on highly active antiretroviral therapy. J Neurovirol 11:180–189 [DOI] [PubMed] [Google Scholar]
  22. Conant K, Garzino-Demo A, Nath A, McArthur JC, Halliday W, Power C, Gallo RC, Major EO (1998) Induction of monocyte chemoattractant protein-1 in HIV-1 Tat-stimulated astrocytes and elevation in AIDS dementia. Proc Natl Acad Sci U S A 95:3117–3121 [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Cosenza MA, Zhao ML, Si Q, Lee SC (2002) Human brain parenchymal microglia express CD14 and CD45 and are productively infected by HIV-1 in HIV-1 encephalitis. Brain Pathol 12:442–455 [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Cosenza-Nashat MA, Bauman A, Zhao ML, Morgello S, Suh HS, Lee SC (2011) Cannabinoid receptor expression in HIV encephalitis and HIV-associated neuropathologic comorbidities. Neuropathol Appl Neurobiol [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Croxford JL, Miller SD (2003) Immunoregulation of a viral model of multiple sclerosis using the synthetic cannabinoid R+WIN55,212. J Clin Invest 111:1231–1240 [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Danbolt NC (2001) Glutamate uptake. Prog Neurobiol 65:1–105 [DOI] [PubMed] [Google Scholar]
  27. D’Aversa TG, Eugenin EA, Berman JW (2005) NeuroAIDS: contributions of the human immunodeficiency virus-1 proteins Tat and gp120 as well as CD40 to microglial activation. J Neurosci Res 81:436–446 [DOI] [PubMed] [Google Scholar]
  28. Ehrhart J, Obregon D, Mori T, Hou H, Sun N, Bai Y, Klein T, Fernandez F, Tan J, Shytle RD (2005) Stimulation of cannabinoid receptor 2 (CB2) suppresses microglial activation. J Neuroinflammation 2:29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Eljaschewitsch E, Witting A, Mawrin C, Lee T, Schmidt PM, Wolf S, Hoertnagl H, Raine CS, Schneider-Stock R, Nitsch R, Ullrich O (2006) The endocannabinoid anandamide protects neurons during CNS inflammation by induction of MKP-1 in microglial cells. Neuron 49:67–79 [DOI] [PubMed] [Google Scholar]
  30. Esposito G, Ligresti A, Izzo AA, Bisogno T, Ruvo M, Di Rosa M, Di Marzo V, Iuvone T (2002) The endocannabinoid system protects rat glioma cells against HIV-1 Tat protein-induced cytotoxicity. Mechanism and regulation. J Biol Chem 277:50348–50354 [DOI] [PubMed] [Google Scholar]
  31. Facchinetti F, Del Giudice E, Furegato S, Passarotto M, Leon A (2003) Cannabinoids ablate release of TNFalpha in rat microglial cells stimulated with lypopolysaccharide. Glia 41:161–168 [DOI] [PubMed] [Google Scholar]
  32. Ferris MJ, Frederick-Duus D, Fadel J, Mactutus C, Booze R Dopamine system vulnerability in HIV-infection: a brief review and initial study using in vivo brain microdialysis in awake, freely moving rats. J Neuroimmune Pharmacol [Google Scholar]
  33. Fine SM, Angel RA, Perry SW, Epstein LG, Rothstein JD, Dewhurst S, Gelbard HA (1996) Tumor necrosis factor alpha inhibits glutamate uptake by primary human astrocytes. Implications for pathogenesis of HIV-1 dementia. J Biol Chem 271:15303–15306 [DOI] [PubMed] [Google Scholar]
  34. Finley MJ, Chen X, Bardi G, Davey P, Geller EB, Zhang L, Adler MW, Rogers TJ (2008) Bi-directional heterologous desensitization between the major HIV-1 co-receptor CXCR4 and the kappa-opioid receptor. J Neuroimmunol 197:114–123 [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Finley MJ, Steele A, Cornwell WD, Rogers TJ (2011) Transcriptional regulation of the major HIV-1 coreceptor, CXCR4, by the {kappa} opioid receptor. J Leukoc Biol [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Galey D, Becker K, Haughey N, Kalehua A, Taub D, Woodward J, Mattson MP, Nath A (2003) Differential transcriptional regulation by human immunodeficiency virus type 1 and gp120 in human astrocytes. J Neurovirol 9:358–371 [DOI] [PubMed] [Google Scholar]
  37. Gannon P, Khan MZ, Kolson DL (2011) Current understanding of HIV-associated neurocognitive disorders pathogenesis. Curr Opin Neurol [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Gekker G, Lokensgard JR, Peterson PK (2001) Naltrexone potentiates anti-HIV-1 activity of antiretroviral drugs in CD4+ lymphocyte cultures. Drug Alcohol Depend 64:257–263 [DOI] [PubMed] [Google Scholar]
  39. Gekker G, Hu S, Wentland MP, Bidlack JM, Lokensgard JR, Peterson PK (2004) Kappa-opioid receptor ligands inhibit cocaine-induced HIV-1 expression in microglial cells. J Pharmacol Exp Ther 309:600–606 [DOI] [PubMed] [Google Scholar]
  40. Gelman BB, Spencer JA, Holzer CE, Soukup VM (2006) Abnormal striatal dopaminergic synapses in national neuroAIDS tissue consortium subjects with HIV encephalitis. J NeuroImmune Pharmacol 2 [DOI] [PubMed] [Google Scholar]
  41. Gendelman HE (ed) (2005) The neurology of AIDS. Oxford University Press, Oxford [Google Scholar]
  42. Ghorpade A, Persidsky Y, Swindells S, Borgmann K, Persidsky R, Holter S, Cotter R, Gendelman HE (2005) Neuroinflammatory responses from microglia recovered from HIV-1-infected and seronegative subjects. J Neuroimmunol 163:145–156 [DOI] [PubMed] [Google Scholar]
  43. Gonzalez-Scarano F, Martin-Garcia J (2005) The neuropathogenesis of AIDS. Nat Rev Immunol 5:69–81 [DOI] [PubMed] [Google Scholar]
  44. Gorantla S, Makarov E, Roy D, Finke-Dwyer J, Murrin LC, Gendelman HE, Poluektova L (2010) Immunoregulation of a CB2 receptor agonist in a murine model of neuroAIDS. J Neuroimmune Pharmacol 5:456–468 [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Hampson AJ, Grimaldi M, Axelrod J, Wink D (1998) Cannabidiol and (−)Delta9-tetrahydrocannabinol are neuroprotective antioxidants. Proc Natl Acad Sci U S A 95:8268–8273 [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Haughey NJ, Cutler RG, Tamara A, McArthur JC, Vargas DL, Pardo CA, Turchan J, Nath A, Mattson MP (2004) Perturbation of sphingolipid metabolism and ceramide production in HIV-dementia. Ann Neurol 55:257–267 [DOI] [PubMed] [Google Scholar]
  47. Heaton RK, Franklin DR, Ellis RJ, McCutchan JA, Letendre SL, Leblanc S, Corkran SH, Duarte NA, Clifford DB, Woods SP, Collier AC, Marra CM, Morgello S, Mindt MR, Taylor MJ, Marcotte TD, Atkinson JH, Wolfson T, Gelman BB, McArthur JC, Simpson DM, Abramson I, Gamst A, Fennema-Notestine C, Jernigan TL, Wong J, Grant I (2011) HIV-associated neurocognitive disorders before and during the era of combination antiretroviral therapy: differences in rates, nature, and predictors. J Neurovirol 17:3–16 [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Howlett AC, Breivogel CS, Childers SR, Deadwyler SA, Hampson RE, Porrino LJ (2004) Cannabinoid physiology and pharmacology: 30 years of progress. Neuropharmacology 47(Suppl 1):345–358 [DOI] [PubMed] [Google Scholar]
  49. Hu S, Ali H, Sheng WS, Ehrlich LC, Peterson PK, Chao CC (1999) Gp-41-mediated astrocyte inducible nitric oxide synthase mRNA expression: involvement of interleukin-1beta production by microglia. J Neurosci 19:6468–6474 [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Itoh K, Mehraein P, Weis S (2000) Neuronal damage of the substantia nigra in HIV-1 infected brains. Acta Neuropathol (Berl) 99:376–384 [DOI] [PubMed] [Google Scholar]
  51. Kaul M, Lipton SA (2006) Mechanisms of neuroimmunity and neurodegeneration associated with HIV-1 infection and AIDS. J Neuroimmune Pharmacol 1:138–151 [DOI] [PubMed] [Google Scholar]
  52. Kaul M, Zheng J, Okamato S, Gendelman HE, Lipton SA (2005) HIV-1 infection and AIDS: consequences for the central nervous system. Cell Death Differ 12:893–904 [DOI] [PubMed] [Google Scholar]
  53. Khurdayan VK, Buch S, El-Hage N, Lutz SE, Goebel SM, Singh IN, Knapp PE, Turchan-Cholewo J, Nath A, Hauser KF (2004) Preferential vulnerability of astroglia and glial precursors to combined opioid and HIV-1 Tat exposure in vitro. Eur J Neurosci 19:3171–3182 [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Kim SY, Li J, Bentsman G, Brooks AI, Volsky DJ (2004) Microarray analysis of changes in cellular gene expression induced by productive infection of primary human astrocytes: implications for HAD. J Neuroimmunol 157:17–26 [DOI] [PubMed] [Google Scholar]
  55. Klegeris A, Bissonnette CJ, McGeer PL (2003) Reduction of human monocytic cell neurotoxicity and cytokine secretion by ligands of the cannabinoid-type CB2 receptor. Br J Pharmacol 139:775–786 [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Klein TW, Cabral GA (2006) Cannabinoid-induced immune suppression and modulation of antigen-presenting cells. J Neuroimmune Pharmacol 1:50–64 [DOI] [PubMed] [Google Scholar]
  57. Klein TW, Newton C, Larsen K, Lu L, Perkins I, Nong L, Friedman H (2003) The cannabinoid system and immune modulation. J Leukoc Biol 74:486–496 [DOI] [PubMed] [Google Scholar]
  58. Koenig S, Gendelman HE, Orenstein JM, Dal Canto MC, Pezeshkpour GH, Yungbluth M, Janotta F, Aksamit A, Martin MA, Fauci AS (1986) Detection of AIDS virus in macrophages in brain tissue from AIDS patients with encephalopathy. Science 233:1089–1093 [DOI] [PubMed] [Google Scholar]
  59. Kosel BW, Aweeka FT, Benowitz NL, Shade SB, Hilton JF, Lizak PS, Abrams DI (2002) The effects of cannabinoids on the pharmacokinetics of indinavir and nelfinavir. AIDS 16:543–550 [DOI] [PubMed] [Google Scholar]
  60. Koutsilieri E, Sopper S, Scheller C, ter Meulen V, Riederer P (2002a) Involvement of dopamine in the progression of AIDS Dementia Complex. J Neural Transm 109:399–410 [DOI] [PubMed] [Google Scholar]
  61. Koutsilieri E, Sopper S, Scheller C, ter Meulen V, Riederer P (2002b) Parkinsonism in HIV dementia. J Neural Transm 109:767–775 [DOI] [PubMed] [Google Scholar]
  62. Lawson LJ, Perry VH, Dri P, Gordon S (1990) Heterogeneity in the distribution and morphology of microglia in the normal adult mouse brain. Neuroscience 39:151–170 [DOI] [PubMed] [Google Scholar]
  63. Lecapitaine NJ, Zhang P, Winsauer P, Walker E, Vande Stouwe C, Porretta C, Molina PE (2011) Chronic delta-9-tetrahydrocannabinol administration increases lymphocyte CXCR4 expression in rhesus macaques. J Neuroimmune Pharmacol [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Lee SC, Dickson DW, Liu W, Brosnan CF (1993) Induction of nitric oxide synthase activity in human astrocytes by interleukin-1 beta and interferon-gamma. J Neuroimmunol 46:19–24 [DOI] [PubMed] [Google Scholar]
  65. Lim SP, Garzino-Demo A (2000) The human immunodeficiency virus type 1 Tat protein up-regulates the promoter activity of the beta-chemokine monocyte chemoattractant protein 1 in the human astrocytoma cell line U-87 MG: role of SP-1, AP-1, and NF-kappaB consensus sites. J Virol 74:1632–1640 [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Lipton SA, Gendelman HE (1995) Seminars in medicine of the Beth Israel Hospital, Boston. Dementia associated with the acquired immunodeficiency syndrome. N Engl J Med 332:934–940 [DOI] [PubMed] [Google Scholar]
  67. Lokensgard JR, Gekker G, Ehrlich LC, Hu S, Chao CC, Peterson PK (1997) Proinflammatory cytokines inhibit HIV-1(SF162) expression in acutely infected human brain cell cultures. J Immunol 158:2449–2455 [PubMed] [Google Scholar]
  68. Lokensgard JR, Gekker G, Peterson PK (2002) Kappa-opioid receptor agonist inhibition of HIV-1 envelope glycoprotein-mediated membrane fusion and CXCR4 expression on CD4(+) lymphocytes. Biochem Pharmacol 63:1037–1041 [DOI] [PubMed] [Google Scholar]
  69. Lu TS, Avraham HK, Seng S, Tachado SD, Koziel H, Makriyannis A, Avraham S (2008) Cannabinoids inhibit HIV-1 Gp120-mediated insults in brain microvascular endothelial cells. J Immunol 181:6406–6416 [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Marsicano G, Moosmann B, Hermann H, Lutz B, Behl C (2002) Neuroprotective properties of cannabinoids against oxidative stress: role of the cannabinoid receptor CB1. J Neurochem 80:448–456 [DOI] [PubMed] [Google Scholar]
  71. Marsicano G, Goodenough S, Monory K, Hermann H, Eder M, Cannich A, Azad SC, Cascio MG, Gutierrez SO, van der Stelt M, Lopez-Rodriguez ML, Casanova E, Schutz G, Zieglgansberger W, Di Marzo V, Behl C, Lutz B (2003) CB1 cannabinoid receptors and on-demand defense against excitotoxicity. Science 302:84–88 [DOI] [PubMed] [Google Scholar]
  72. Mattson MP, Haughey NJ, Nath A (2005) Cell death in HIV dementia. Cell Death Differ 12(Suppl 1):893–904 [DOI] [PubMed] [Google Scholar]
  73. McArthur JC, Brew BJ, Nath A (2005) Neurological complications of HIV infection. Lancet Neurol 4:543–555 [DOI] [PubMed] [Google Scholar]
  74. Minghetti L, Visentin S, Patrizio M, Franchini L, Ajmone-Cat MA, Levi G (2004) Multiple actions of the human immunodeficiency virus type-1 Tat protein on microglial cell functions. Neurochem Res 29:965–978 [DOI] [PubMed] [Google Scholar]
  75. Molina PE, Winsauer P, Zhang P, Walker E, Birke L, Amedee A, Stouwe CV, Troxclair D, McGoey R, Varner K, Byerley L, Lamotte L (2010) Cannabinoid administration attenuates the progression of simian immunodeficiency virus. AIDS Res Hum Retroviruses [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Mollace V, Nottet HS, Clayette P, Turco MC, Muscoli C, Salvemini D, Perno CF (2001) Oxidative stress and neuroAIDS: triggers, modulators and novel antioxidants. Trends Neurosci 24:411–416 [DOI] [PubMed] [Google Scholar]
  77. Nagayama T, Sinor AD, Simon RP, Chen J, Graham SH, Jin K, Greenberg DA (1999) Cannabinoids and neuroprotection in global and focal cerebral ischemia and in neuronal cultures. J Neurosci 19:2987–2995 [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Nath A, Sacktor N (2006) Influence of highly active antiretroviral therapy on persistence of HIV in the central nervous system. Curr Opin Neurol 19:358–361 [DOI] [PubMed] [Google Scholar]
  79. Nath A, Anderson C, Jones M, Maragos W, Booze R, Mactutus C, Bell J, Hauser KF, Mattson M (2000) Neurotoxicity and dysfunction of dopaminergic systems associated with AIDS dementia. J Psychopharmacol 14:222–227 [DOI] [PubMed] [Google Scholar]
  80. Noe SN, Nyland SB, Ugen K, Friedman H, Klein TW (1998) Cannabinoid receptor agonists enhance syncytia formation in MT-2 cells infected with cell free HIV-1MN. Adv Exp Med Biol 437:223–229 [DOI] [PubMed] [Google Scholar]
  81. Palazuelos J, Aguado T, Pazos MR, Julien B, Carrasco C, Resel E, Sagredo O, Benito C, Romero J, Azcoitia I, Fernandez-Ruiz J, Guzman M, Galve-Roperh I (2009) Microglial CB2 cannabinoid receptors are neuroprotective in Huntington’s disease excitotoxicity. Brain 132:3152–3164 [DOI] [PubMed] [Google Scholar]
  82. Paul RH, Brickman AM, Navia B, Hinkin C, Malloy PF, Jefferson AL, Cohen RA, Tate DF, Flanigan TP (2005) Apathy is associated with volume of the nucleus accumbens in patients infected with HIV. J Neuropsychiatry Clin Neurosci 17:167–171 [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Persidsky Y, Ghorpade A, Rasmussen J, Limoges J, Liu XJ, Stins M, Fiala M, Way D, Kim KS, Witte MH, Weinand M, Carhart L, Gendelman HE (1999) Microglial and astrocyte chemokines regulate monocyte migration through the blood-brain barrier in human immunodeficiency virus-1 encephalitis. Am J Pathol 155:1599–1611 [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Peterson PK, Hu S, Anderson WR, Chao CC (1994) Nitric oxide production and neurotoxicity mediated by activated microglia from human versus mouse brain. J Infect Dis 170:457–460 [DOI] [PubMed] [Google Scholar]
  85. Peterson PK, Gekker G, Lokensgard JR, Bidlack JM, Chang AC, Fang X, Portoghese PS (2001) Kappa-opioid receptor agonist suppression of HIV-1 expression in CD4+ lymphocytes. Biochem Pharmacol 61:1145–1151 [DOI] [PubMed] [Google Scholar]
  86. Peterson PK, Gekker G, Hu S, Cabral G, Lokensgard JR (2004) Cannabinoids and morphine differentially affect HIV-1 expression in CD4(+) lymphocyte and microglial cell cultures. J Neuroimmunol 147:123–126 [DOI] [PubMed] [Google Scholar]
  87. Pocernich CB, Sultana R, Mohmmad-Abdul H, Nath A, Butterfield DA (2005) HIV-dementia, Tat-induced oxidative stress, and antioxidant therapeutic considerations. Brain Res Brain Res Rev 50:14–26 [DOI] [PubMed] [Google Scholar]
  88. Ponti W, Rubino T, Bardotti M, Poli G, Parolaro D (2001) Cannabinoids inhibit nitric oxide production in bone marrow derived feline macrophages. Vet Immunol Immunopathol 82:203–214 [DOI] [PubMed] [Google Scholar]
  89. Price DA, Martinez AA, Seillier A, Koek W, Acosta Y, Fernandez E, Strong R, Lutz B, Marsicano G, Roberts JL, Giuffrida A (2009) WIN55,212–2, a cannabinoid receptor agonist, protects against nigrostriatal cell loss in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mouse model of Parkinson’s disease. Eur J Neurosci 29:2177–2186 [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Raborn ES, Cabral GA (2010) Cannabinoid inhibition of macrophage migration to the trans-activating (Tat) protein of HIV-1 is linked to the CB(2) cannabinoid receptor. J Pharmacol Exp Ther 333:319–327 [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Rock RB, Hu S, Deshpande A, Munir S, May BJ, Baker CA, Peterson PK, Kapur V (2005) Transcriptional response of human microglial cells to interferon-gamma. Genes Immun 6:712–719 [DOI] [PubMed] [Google Scholar]
  92. Rock RB, Gekker G, Hu S, Sheng WS, Cabral GA, Martin BR, Peterson PK (2007) WIN55,212–2-mediated inhibition of HIV-1 expression in microglial cells: involvement of cannabinoid receptors. J Neuroimmune Pharmacol 2:178–183 [DOI] [PubMed] [Google Scholar]
  93. Roth MD, Tashkin DP, Whittaker KM, Choi R, Baldwin GC (2005) Tetrahydrocannabinol suppresses immune function and enhances HIV replication in the huPBL-SCID mouse. Life Sci 77:1711–1722 [DOI] [PubMed] [Google Scholar]
  94. Sardar AM, Czudek C, Reynolds GP (1996) Dopamine deficits in the brain: the neurochemical basis of parkinsonian symptoms in AIDS. Neuroreport 7:910–912 [DOI] [PubMed] [Google Scholar]
  95. Shen M, Thayer SA (1998) Cannabinoid receptor agonists protect cultured rat hippocampal neurons from excitotoxicity. Mol Pharmacol 54:459–462 [DOI] [PubMed] [Google Scholar]
  96. Sheng WS, Hu S, Hegg CC, Thayer SA, Peterson PK (2000) Activation of human microglial cells by HIV-1 gp41 and Tat proteins. Clin Immunol 96:243–251 [DOI] [PubMed] [Google Scholar]
  97. Sheng WS, Hu S, Lokensgard JR, Peterson PK (2003) U50,488 inhibits HIV-1 Tat-induced monocyte chemoattractant protein-1 (CCL2) production by human astrocytes. Biochem Pharmacol 65:9–14 [DOI] [PubMed] [Google Scholar]
  98. Sheng WS, Hu S, Min X, Cabral GA, Lokensgard JR, Peterson PK (2005) Synthetic cannabinoid WIN55,212–2 inhibits generation of inflammatory mediators by IL-1beta-stimulated human astrocytes. Glia 49:211–219 [DOI] [PubMed] [Google Scholar]
  99. Sheng WS, Hu S, Herr G, Ni HT, Rock RB, Gekker G, Lokensgard JR, Peterson PK (2007) Human neural precursor cells express functional kappa-opioid receptors. J Pharmacol Exp Ther 322:957–963 [DOI] [PubMed] [Google Scholar]
  100. Sheng WS, Hu S, Ni HT, Rock RB, Peterson PK (2009) WIN55,212–2 inhibits production of CX3CL1 by human astrocytes: involvement of p38 MAP kinase. J Neuroimmune Pharmacol 4:244–248 [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Shohami E, Mechoulam R (2000) Dexanabinol (HU-211): a nonpsychotropic cannabinoid with neuroprotective properties. Drug Dev Res 50:211–215 [Google Scholar]
  102. Silvers JM, Aksenov MY, Aksenova MV, Beckley J, Olton P, Mactutus CF, Booze RM (2006) Dopaminergic marker proteins in the substantia nigra of human immunodeficiency virus type 1-infected brains. J Neurovirol 12:140–145 [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Srivastava MD, Srivastava BI, Brouhard B (1998) Delta9 tetrahydrocannabinol and cannabidiol alter cytokine production by human immune cells. Immunopharmacology 40:179–185 [DOI] [PubMed] [Google Scholar]
  104. Stefano GB, Salzet M, Bilfinger TV (1998a) Long-term exposure of human blood vessels to HIV gp120, morphine, and anandamide increases endothelial adhesion of monocytes: uncoupling of nitric oxide release. J Cardiovasc Pharmacol 31:862–868 [DOI] [PubMed] [Google Scholar]
  105. Stefano GB, Salzet M, Rialas CM, Mattocks D, Fimiani C, Bilfinger TV (1998b) Macrophage behavior associated with acute and chronic exposure to HIV GP120, morphine and anandamide: endothelial implications. Int J Cardiol 64(Suppl 1):S3–S13 [DOI] [PubMed] [Google Scholar]
  106. Su ZZ, Kang DC, Chen Y, Pekarskaya O, Chao W, Volsky DJ, Fisher PB (2002) Identification and cloning of human astrocyte genes displaying elevated expression after infection with HIV-1 or exposure to HIV-1 envelope glycoprotein by rapid subtraction hybridization, RaSH. Oncogene 21:3592–3602 [DOI] [PubMed] [Google Scholar]
  107. Su ZZ, Kang DC, Chen Y, Pekarskaya O, Chao W, Volsky DJ, Fisher PB (2003) Identification of gene products suppressed by human immunodeficiency virus type 1 infection or gp120 exposure of primary human astrocytes by rapid subtraction hybridization. J Neurovirol 9:372–389 [DOI] [PubMed] [Google Scholar]
  108. Timpone JG, Wright DJ, Li N, Egorin MJ, Enama ME, Mayers J, Galetto G (1997) The safety and pharmacokinetics of single-agent and combination therapy with megestrol acetate and dronabinol for the treatment of HIV wasting syndrome. The DATRI 004 Study Group. Division of AIDS Treatment Research Initiative. AIDS Res Hum Retroviruses 13:305–315 [DOI] [PubMed] [Google Scholar]
  109. Turchan-Cholewo J, Liu Y, Gartner S, Reid R, Jie C, Peng X, Chen KC, Chauhan A, Haughey N, Cutler R, Mattson MP, Pardo C, Conant K, Sacktor N, McArthur JC, Hauser KF, Gairola C, Nath A (2006) Increased vulnerability of ApoE4 neurons to HIV proteins and opiates: protection by diosgenin and L-deprenyl. Neurobiol Dis 23:109–119 [DOI] [PubMed] [Google Scholar]
  110. United Nations Program on HIV/AIDS (UNAIDS)/World Health Organization (2010) 2010 Report on the global AIDS epidemic, in, UNAIDS, Geneva [Google Scholar]
  111. Wallace DR, Dodson S, Nath A, Booze RM (2006) Estrogen attenuates gp120- and tat1–72-induced oxidative stress and prevents loss of dopamine transporter function. Synapse 59:51–60 [DOI] [PubMed] [Google Scholar]
  112. Walter L, Franklin A, Witting A, Wade C, Xie Y, Kunos G, Mackie K, Stella N (2003) Nonpsychotropic cannabinoid receptors regulate microglial cell migration. J Neurosci 23:1398–1405 [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Wang Z, Pekarskaya O, Bencheikh M, Chao W, Gelbard HA, Ghorpade A, Rothstein JD, Volsky DJ (2003) Reduced expression of glutamate transporter EAAT2 and impaired glutamate transport in human primary astrocytes exposed to HIV-1 or gp120. Virology 312:60–73 [DOI] [PubMed] [Google Scholar]
  114. Wang GJ, Chang L, Volkow ND, Telang F, Logan J, Ernst T, Fowler JS (2004) Decreased brain dopaminergic transporters in HIV-associated dementia patients. Brain 127:2452–2458 [DOI] [PubMed] [Google Scholar]
  115. Wiley CA, Schrier RD, Nelson JA, Lampert PW, Oldstone MB (1986) Cellular localization of human immunodeficiency virus infection within the brains of acquired immune deficiency syndrome patients. Proc Natl Acad Sci U S A 83:7089–7093 [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Zhou BY, Liu Y, Kim B, Xiao Y, He JJ (2004) Astrocyte activation and dysfunction and neuron death by HIV-1 Tat expression in astrocytes. Mol Cell Neurosci 27:296–305 [DOI] [PubMed] [Google Scholar]

RESOURCES