Abstract
Methamphetamine (Meth) is an addictive psychostimulant widely abused around the world. The chronic use of Meth produces neurotoxicity featured by dopaminergic terminal damage and microgliosis, resulting in serious neurological and behavioral consequences. Ample evidence indicate that Meth causes microglial activation and resultant secretion of pro-inflammatory molecules leading to neural injury. However, the mechanisms underlying Meth-induced microglial activation remain to be determined. In this review, we attempt to address the effects of Meth on human immunodeficiency virus (HIV)-associated microglia activation both in vitro and in-vivo. Meth abuse not only increases HIV transmission but also exacerbates progression of HIV-associated neurocognitive disorders (HAND) through activation of microglia. In addition, the therapeutic potential of anti-inflammatory drugs on ameliorating Meth-induced microglia activation and resultant neuronal injury is discussed.
Keywords: Drug abuse, methamphetamine, microglia, neuroinflammation, neurotoxicity
Introduction
Methamphetamine (Meth) is a highly-abused psychostimulant and the second most widely used illicit drug worldwide (after cannabis) [1]. Chronic abuse eventually leads to the neurotoxic regimen, which induces the psychological and behavioral abnormalities, such as increased aggressive behavior and craving for the drug [2-5]. The long-term neurotoxic effects of Meth are well established by neuroimaging studies and psychological tests and confirmed in both rodents and non-human primates [6-8]. Although the primary target of Meth is dopaminergic terminal, the neuropathological changes are not only limited in the striatum. A broader scope of the neural injury has been observed in human subjects with a decreased volume of the hippocampus and the hypertrophy in white matter [9]. Repeated administration of Meth also impaired cognitive function, which could be partially explained by current dopamine-based neurotoxic mechanisms [10-13], suggesting other mechanisms may be involved in Meth-associated neuropathology. Increasing evidence indicate that neuroinflammation featured by microglial activation plays an important role in Meth-induced neurotoxicity (Figure 1). The notion is supported by recent studies that anti-inflammatory drug ibudilast attenuated Meth dependence and Meth-induced neural injury [14-17].
Figure 1.
Schematic diagrams illustrating the roles played by microglia in health and disease (Meth abuse) conditions. In health, microglia activation is restricted by neuronal immunosuppressive signals. In turn, the microglia secretes neurotrophic factors to support the neuronal function. In Meth abuse condition (disease), however, Meth causes neuronal cell damage, especially the dopaminergic neuronal terminals. The damaged neurons release danger-associated signals leading to microglia activation. The activated microglia produces proinflammatory substances (neurotoxins), exacerbating Meth-associated neurotoxic activity. Thus, inhibition of microglia activation and resultant production of proinflammatory substances could be an important strategy for therapeutic intervention.
Studies have shown that microgliosis is an early response to Meth abuse and such a response lasts for a long time even after abstinence [18,19]. Over-activated microglia are found in multiple brain regions in individuals with Meth abuse, but not in those with cocaine use [19,20]. The time-course, dose-response, and pharmacological profiles of Meth-induced microglial activation indicate that over-activated microglia are not merely a response subsequent to nerve terminal damage, but a specific pharmacological marker of Meth-induced neurotoxicity [18,21]. As microglia are regulated by a variety of inhibitory signals such as CX3CL1, CD200, CD22, or CD172 [22-24], the impact of repeated Meth administration on inhibitory signaling molecules in central nervous system (CNS) were investigated as the potential therapeutic targets [25,26]. On the other hand, since the release of neurotoxins including pro-inflammatory cytokines and super oxidative factors are the primary neuronal toxic mediators of microglia [27], the production of these pro-inflammatory mediators after Meth treatment was analyzed [28]. However, the results on the immune modulatory effects of Meth are inconsistent. While its suppressive role was reported in most studies in the peripheral immune system [29-32], Meth was primarily considered as a pro-inflammatory mediator in the CNS [33-35]. A recent study on cultured microglia showed that Meth had limited impacts on microglia production of proinflammatory cytokines indicating the biological and molecular intricacy of this drug [36]. Further investigations on potential roles of the other CNS-specific factors and neuronal danger-associated molecular patterns (DAMPs) are needed. Meth dependence is one of the most common co-morbid conditions among the HIV-infected population [37]. In comparison with the independent effect of Meth abuse or HIV infection, the combined action of HIV infection and Meth dependence resulted in more severe impairment on neurocognition [38-40]. Studies on the interactions between Meth and HIV proteins in animal models have demonstrated their synergistic effects on cognitive deficits [40,41] and altered behaviors [42,43]. However, the precise mechanisms underlying Meth exacerbation of HAND remain unclear. It has been shown that Meth-taking patients with HIV encephalitis (HIVE) exhibited a significant microgliosis, but not astrogliosis [44], suggesting that enhanced microglial activation underlies the cross-talk of HIV-1 infection and Meth dependence. Moreover, Meth abuse enhances opportunistic microbial brain infection [45,46] and increases lipopolysaccharide (LPS)-mediated human macrophage production of proinflammatory cytokines [47]. LPS was found to potentiate Meth-associated neurotoxicity [48]. Based on these results, whether chronic Meth abuse could enhance the pre-existing neuroinflammation and promote the progression of another comorbid disease are emerging topics that are worthy of further investigation.
Acute and chronic consequences of Meth abuse
Meth is a synthetic lipophilic drug that is blood brain barrier (BBB) permeable [49]. It derived from amphetamine, but with higher binding affinity to monoamine transporters [50]. Meth administration induces a remarkable outflow of monoamines from synaptic vesicles to cytosol on presynaptic neuron [51-53]. By reverse transportation and freely diffusion, Meth induces potent efflux of dopamine to postsynaptic site causing strong and long-lasting euphoric effect. A comparable dopamine dynamic study suggested that single injection of Meth on the concentration of 2.5 mg/kg induced around 5 times of dopamine release compared to cocaine on 40 mg/kg [54]. Because it is easy to synthesize and is much more potent on psychostimulant effect than other stimulants, Meth has already widely abused all around the world.
The instant effects of Meth include increased attention, activity, and wakefulness; decreased fatigue and appetite; euphoria and rush experience. Hyperthermia and irregular heartbeat are two most prominent toxic effects of hyperthermia and irregular heartbeat overdose [49]. On the other hand, chronic use of Meth induces a series of negative consequences including addiction, anxiety, confusion, insomnia, mood disturbances, and violent behaviors [55]. In addition to these psychotic symptoms, chronic abusers are often associated with cognitive deficits ranging from impaired pulse control, working memory and decision-making [56-58]. These symptoms are accompanied by the Meth-induced neuropathological changes including the damage to dopamine and serotonin axons, loss of gray matter, hypertrophy of the white matter, and microgliosis. Neuroimaging studies with specific probes implicated a monoamine transporter reduction and dopaminergic terminal degeneration [59,60]. Moreover, magnetic resonance imaging (MRI) studies indicated that Meth-associated neurodegeneration was not restricted to the striatum [9,61]. Considerable shrinkage of hippocampi, gray matter, cingulate cortex, limbic cortex and paralimbic cortex was observed in recreational abusers [9]. Multiple neurotoxic events are associated with Meth abuse including oxidative stress, excitotoxicity, hyperthermia, mitochondrial dysfunction, endoplasmic reticulum stress, and neuroinflammatory responses. Here, we primarily focus on neuroinflammation associated with microglial activation.
Microgliosis: a neurotoxic marker of Meth addiction
Microglial activation is characterized by proliferation, morphologically change, migration and inflammatory secretion profiles [62]. It is well-established that the activation of microglia is relative to neurodegenerative diseases, brain injury and toxicant-induced damage to the CNS [63]. In animal model injected with Meth, microgliosis was found in multiple brain areas [64]. A human study validated the remarkable microglial activation in all brain regions using positron emission tomography (PET) with a specific radiotracer [19]. The dose- and time-response of Meth-induced microglial activation performed on Meth-administrated rat indicated that microglial response preceded both terminal neuronal degeneration and astrocyte activation [18]. As it is difficult to differentiate infiltrated monocyte/macrophage with activated microglia by immunohistochemistry staining, the possibility that Meth increases the trafficking of peripheral immune cells into the brain needs to be further proved. The irradiated mice were rescued with bone marrow transplantation from “green mice”, a transgenic mouse line with an enhanced GFP (eGFP) expression in all tissues except for erythrocytes and hair. To determine if Meth increases the peripheral monocyte infiltration, the peripheral monocytes in the brain with eGFP were detected after Meth administration. Two days after four injections of neurotoxic regimen of Meth (5 mg/kg) and/or physiological saline in with a 2-h interval, the brains were harvested, fixed and the sectioned. The stratum sections were examined under fluorescent microscope. While the resident microglia were significantly activated in the striatum, no infiltrated eGFP-expressing cells (migrated hematopoietic cells) were detected [25]. Although this study suggests that there is no evidence of transmigration of peripheral hematopoietic cells cross the BBB in response to Meth administration, the role of BBB impairment in Meth-induced neuroinflammation is still in debate. The alteration of BBB permeability was found shortly after Meth administration and lasted long in multiple brain areas including stratum, amygdala, and hippocampus [65,66]. Moreover, Meth application was found to induce the oxidative stress in cultured primary human brain microvascular endothelial cells (BMVEC) [67]. The impairment of barrier function of BBB was determined both in vivo using fluorescent tracer and in vitro via transendothelial electrical resistance (TEER) test. The application of Meth diminished the tightness of BMVEC monolayers by decreasing the expression of cell membrane-associated tight junction proteins and thus, enhanced the monocyte transendothelial migration [67]. The increased monocyte passage through the endothelial cells monolayer can be blocked by a specific inhibitor of Arp 2/3 complex, indicating that actin cytoskeletal dynamics play an important role in Meth-induced transendothelial monocyte migration [68]. Taken together, although the Meth-induced microglial activation is validated in both animal models and human abusers, it is still controversial whether Meth-associated inflammatory response is mediated by the resident microglia other than the peripheral immune cells infiltration.
To determine the mechanism of microglial activation, various experimental groups were designed to mimic selected pharmacological elements of Meth action [21]. As another psychostimulant with high binding affinity with plasma dopamine transporter, cocaine failed to trigger microglial activation in Meth administrated rat [21]. Also, the administration of cocaine was found less relative to long-term neurotoxicity [20,69]. The involvement of dopamine receptors in Meth-induced microglial activation was also evaluated. However, neither D1 nor D2 receptor agonist replicated Meth-induced microglial activation, which suggested that microglial activation was independent of Meth-induced release of dopamine [21]. Furthermore, the levorotary enantiomer of Meth (L-Meth), a molecular sharing the most receptor targets with Meth but with much less binding affinities, failed to activate microglia. Because L-Meth is much less neurotoxic compared to dextrorotary enantiomer [70], its failure in inducing microglial activation further suggests that Meth-induced neuroinflammation is an essential event in the Meth neurotoxic cascade. On the other hand, the factors that mitigate neurotoxicity of Meth such as lower ambient temperature and NMDA receptor antagonists also reduce microgliosis along with their neuroprotective effects [18,71]. Moreover, the tolerance of Meth toxicity is also associated with attenuated microglial activation. After a neurotoxic challenge with Meth, tolerance was developed to the subsequent neurotoxic effects of Meth [72-74]. Although the second Meth stimulation induced the same extent of hyperthermia and astrocyte activities, microglial activation was blunted [75,76]. Thus, attenuated microglial activation was considered as an important mechanism underlying the reduced Meth-induced neurotoxicity. In contrast, classical immunogen LPS administration significantly increased the microglial activation and potentiated the Meth-induced neurotoxicity [48,77]. While the astrocytes remain reactive even 30 days after Meth administration, microglial activation subsides within 7 days, which was more reflecting the acute Meth-induced neurotoxicity [76]. Based aforementioned evidence, microglial activation could be an important indicator of Meth-induced toxicity.
Although in vivo experiments demonstrate a solid link between neurotoxicity and microglia-mediated neuroinflammation [78], limited studies have been done on Meth effects on cultured microglia. While Meth receptors on microglia remain to be determined, it is unclear whether Meth-induced microglial activation is a receptor-mediated effect. Therefore, testing enantiomers on isolated microglia may, at least in part, address this question because of the different binding affinities of the dextrorotatory/levorotary forms to certain receptors. Since activated microglial primarily contribute to neuronal injury by releasing various neurotoxic factors, many studies focus on the activation of inflammatory signaling pathway and subsequent production of neurotoxins after Meth exposure. A latest study evaluated mRNA levels of inflammatory-associated genes on cultured microglia stimulated by various concentrations of Meth [36]. Proinflammatory cytokines upregulated in vivo were found unchanged in Meth-treated microglial cultures [36]. The inflammatory responses on both whole brain microglia and isolated striatum microglia are consistent, which means there is no regional specificity of microglia activation. Moreover, the IL-1β level and cell viability were found decreased after administration of 1 mM Meth [36]. This was consistent with previous experiments on cultured microglia, which showed the apoptotic effect of Meth in 500 μM, and could be reversed by TNFα and IL-6 through IL-6 receptor and JAK-STAT3 pathway [79]. While most of the studies indicate that there is less possibility for Meth to stimulate the pro-inflammatory response directly, a study performed on CHME-5 cell line (Human fetal microglial immortalized with large T antigen of simian virus 40) suggested that Meth could reactivate HIV transcription in an NF-κB-dependent manner. The NF-κB reporter assay (Luciferase System) and the p65 ELISA of nuclear extracts were used to examine the activity of this cellular transcription factors. The activation of NF-κB was observed in CHME-5 cell treated with 500 µM Meth. The activation effect of Meth started as early as 0.5 h and lasted for 24 h. Moreover, the IκB dominant-negative construct, which lacked the phosphorylation site and could not be dissociated from NF-κB, blocked the Meth-induced activation [80]. It is worth to note that the concentration of Meth used in those experiments was more than 500 µM, which was too high to represent the condition of long-term recreational users. On the other hand, the activation of microglia might also be a consequence of the neuronal release of DAMPs that stimulated by application of Meth. High mobility group box-1 (HMGB1) was found upregulated after Meth administration and mediated the neuroinflammatory response in multiple brain areas [36]. Another candidate neuronal DAMP that might mediate Meth-induced microglial activation is DA-quinones (DAQ). The excess outflow of dopamine induced by Meth could be self-oxidized to DAQ that might play an obligatory role in various Meth-induced neurotoxic effects [81,82], because researchers found DAQ causes a time-dependent activation of cultured microglia. Gene expression study analyzed 101 genes alteration, in which inflammation cytokines, chemokines, and prostaglandins were upregulated, whereas protective neuronal genes were downregulated [81]. The critical roles of dopamine and its oxidative form in Meth-induced microglial activation were further demonstrated by the disruption of DA release from the newly synthesized pool in vivo, which abrogated the microgliosis [83]. Thus, the excessive release of dopamine from vesicles and outflow outside neuronal terminals might function as neuronal DAMPs that could be sensed by regional microglia and initiates the neurotoxic signal cascades [83]. Despite the aforementioned progresses the patterns and mechanisms of microglia activation in the brain need to be further investigated.
The microglia neurotoxicity was initially demonstrated in vitro with primary cultured microglia. After challenge with proinflammatory stimuli such as LPS, IFNβ, or TNFα, the supernatants were transferred to cultured neurons, and the neuronal apoptosis was observed [84]. Ever since these experiments, it has been well established that the toxic microglial secretory products are the major mediators of microglia-induced neurotoxicity [85-88]. For this reason, it is critical to examine whether the proinflammatory factors, released by microglia in response to Meth, are toxic to neurons. It was shown that microglia-associated factors, for example, IL-1α, IL-6, CCL2, and TNFα, were upregulated in mice with a single low-dose regimen of Meth [89]. The microglial-mediated proinflammatory responses were attenuated by minocycline, a selective inhibitor of microglial activation [89]. However, the experiment using mice with genetic deficiency in IL-1α, IL-6, and CCL2 did not show neuroprotection against Meth [89]. Only the mice lack of the TNF1/2 receptors showed attenuated neurotoxicity, indicating that TNF-α is a critical factor in Meth-induced neurotoxicity. Thereby, it is hypothesized that the failure of minocycline in neuroprotection against Meth-associated neuronal damages could be attributed to its incomplete inhibition of TNF-α signaling pathway [89]. Currently, neuroprotective effects targeting microglial activation on Meth-related neurotoxicity in an animal model is controversial [89-92]. The disparity between these mixed results probably attribute to the differences in the dosing regimen or the time of anti-inflammatory drug administration. However, lack of specific selectivity against downstream pathway(s) might also contribute to these ambiguous results. Further studies on identification and inhibition of more specific molecular targets mediating microglia-mediated neuroinflammation are imperative.
Microglia: a potential intersecting point for Meth and HIV
Despite the successfulness of highly active antiretroviral therapy (HAART), 50% of the HIVinfected individuals still suffer from HIV-associated neurological disorder (HAND) [93]. The persistent neuroinflammation was found in HAND patients, even in the HAART era [94-97]. A neuroimaging study revealed that the degree of neuroinflammation marked by the activated microglia inversely correlated with the executive function even in the patient receiving HAART [98]. Meth abuse is prevalent among HIV-infected patients, and the interaction between Meth abuse and HIV pathogenesis is an interesting research topic for many neurovirologists [37,99,100]. In comparison to their independent effects, Meth exposure combined with HIV infection produces severer impairment on neurocognition [38,39]. Studies on the interactions between HIV proteins and Meth in animal models have further demonstrated their synergistic effects on cognitive dysfunctions [40,41] and altered behaviors [42,43]. However, the precise mechanisms for Meth exacerbation of HAND are still not fully understood. More significant microgliosis was observed in Meth-taking patients with HIVE, while astrogliosis was not changed [44]. Thus, enhanced microglial activation could be the intersecting point for the cross-talk of HIV infection and Meth dependence. Because neurons are refractory to the HIV infection, the activated microglia are most likely the contributors to neuronal damages and cognitive dysfunctions due to the release of neurotoxic substances [101]. On the one hand, Meth is associated with increased entry of virus into CNS system, which could be attributed to several potential mechanisms including increased peripheral viral load [102], reduced adherence to antiretroviral therapy [103], and disruption of the blood-brain barrier (BBB) [65,104]. Furthermore, Meth can regulate the HIV replication by targeting HIV-associated cellular factors [105,106]. The potential of dopamine to promote HIV replication is corroborated in rat and monkey models with administration of L-DOPA [107,108]. However, the dopamine concentrations used in the above studies were very high; it is questionable whether such high dopamine concentrations could be found in Meth abusers [109].
Recently, common molecular targets for Meth and HIV intersecting effects gain more attention. It has been suggested that voltage-gated potassium channels (Kv) are involved with neuronal damage and microglial function [110]. HIV protein Tat and gp120 were reported to increase the outward potassium current and proinflammatory cytokines release, resulting in neurotoxic activity [111,112]. The potassium channels on microglia were also associated with Meth neurotoxicity [113]. Thus, it is worth to investigate further whether Meth application could potentiate the Tat- or gp120-induced microglial inflammatory response through specific potassium channels. In addition, some microglia-associated proinflammatory cytokines that are crucial for the progression of Meth- and HIV-induced neuropathological changes are also needed to be further investigated [114-117]. The enhanced TNFα secreted by microglia after Meth stimulation might be responsible for the increased HIV replication [118], NMDA receptor neurotoxicity [119], and BBB disruption [120]. Likewise, upregulated IL-1β and IL-8 expression may also contribute to the Meth and HIV-associated neurotoxic activities [116,117] as demonstrated by their compromising effects on Long-term potentiation (LTP) and cognitive function [121,122].
Meth and neuronal immunosuppressive signals
The neuronal cells are vulnerable to the potential detrimental immune reactions. Except for a few limited brain areas, most neurons are incompetents in regenerating themselves [123]. Thus, in addition to the physical isolation by blood brain barrier, neurons express multiple immune suppressive signals to provide a restricted and immunosuppressed microenvironment, which rapidly turns down uncontrolled microglial activation to prevent secondary neuronal damage [22]. Those signals may be secreted by neurons including TGF-β, CX3CL1, and CD22 or expressed on the membrane such as CD200 and CD47. Among these suppressive signals exchanging between the neuron and glial cells, CX3CL1-CX3CR1 and CD200-CD200R are two most well-studied axes in Meth-associated neuronal dysfunction [25,26]. Ample evidence shows CX3CL1 is the most prominent neuron-derived signal that restricts microglial activation after harmful environmental stimulization [124]. Mice homozygous with CX3CR1-deficient have been widely studied in various neurodegenerative models. Mice with deficient CX3CL1-CXCR1 signaling showed an enhanced microglial activation, resulting in greater neurotoxicity [125]. In Meth-induced neurotoxicity, while the genes of many inflammatory molecules were found upregulated, the expression levels of CX3CR1 and CX3CL1 did not change [126]. Unlike the hypersensitivity of the neurons to MPTP-induced neurotoxicity in a CX3CR1 knockout mouse model, the level of Meth-induced hyperthermia and dopaminergic terminal damage were not enhanced in CX3CR1 deficient mice [25]. It is still unclear, however, if the overexpressed exogenous CX3CL1 could downregulate Meth-induced microglial activation. Thus, although current evidence supports that microglial activation and neuronal damage induced by Meth are not mediated by CX3CR1 signaling, the therapeutic potential of this signaling axis needs further investigation.
Another well-studied neuronal immunosuppressive signal in Meth-induced neurotoxicity is CD200. The neuroprotective roles of CD200 against microglia-induced neurotoxicity were reported in multiple neurodegenerative disease models [127,128]. Consistently, CD200 exerts protective effects on Meth neurotoxicity via decreasing microglial activation [26]. The Fc region of CD200 (CD200-Fc) was tested on neuron-microglia co-culture system and on rats before Meth treatment [26]. CD200-Fc has been proved in vitro that it suppressed the microglial activation and secretion of inflammatory cytokines (IL-1β and TNFα) [26]. Meanwhile, the Meth-induced striatal neurotoxicity was also attenuated by the CD200-Fc application [26]. However, the Meth-associated microglial activation and neuronal damage were not completely blocked by CD200-Fc, indicating the insufficiency of the solo neuronal suppressive signal. Also, because Meth-induced neuronal damages are consisted of multifactorial and complicated processes, the inhibition of microglial activation might not be enough to reverse the toxicity of Meth completely. Lastly, it is still challenging to deliver the antibody drugs into the specific brain areas. Thus, improved delivery systems must facilitate developing the immunosuppressive neuronal signals administration as a therapeutic strategy [129].
Therapeutic studies of Meth-induced neural injury
Currently, the emerging cures for Meth addiction are cognitive behavioral therapy and inpatient treatment. However, the therapeutic effect is barely satisfied. Only 33% of Meth users finished 16 weeks of outpatient counseling and merely 45% patients achieved 3 weeks of Meth abstinence [130]. Even inpatient treatment can reach only 30% long-term abstinent [131]. The reason for this high relapse rate is the Meth-induced neuropsychiatric impairments [132,133]. Long-term abuse of Meth causes structural damages to multiple brain areas followed by the impairment of the cognitive and psychiatric functions [134-136]. The Meth abstainers with brain dysfunctions are more likely to relapse [3,58]. This feedback loop is the major obstacle to the current therapy for Meth dependence. However, with limited successfulness of drug development based on neurotransmitter systems, alternative therapeutic strategies are needed. Because the neuroinflammation plays a critical role in Meth-induced neurotoxicity, overactivated microglia seems to be a promising target for therapeutic approaches.
In recent years, exciting progresses have been made on developing an anti-inflammation strategy against microglia-mediated neuronal damages [137,138]. Minocycline is the most lipophilic tetracycline antibiotic that is proved for an anti-inflammatory effect through inhibition of key inflammatory enzymes, like inducible nitric oxide synthase (iNOS) [139], Matrix metalloproteinases (MMPs) [140], cyclooxygenase-2 (COX2) [141] and Phospholipase A2 (PLA2) [142]. Minocycline blocked the microglial activation and attenuated the Meth-induced neurotoxicity [90,143,144]. After pretreatment of minocycline (40 mg/kg), the behavioral sensitization induced by repeated administration of Meth (3 mg/kg/day) was significantly attenuated [143]. Furthermore, the reduction of dopamine and dopamine transporter (DAT) after Meth are also rescued by application of minocycline [143]. A study performed on monkeys further confirmed the neuroprotective effect of minocycline against Meth [90]. A reduction of DAT after repeated administration of Meth (2 mg/kg) was observed by PET, with or without minocycline (200 mg). The treatment of minocycline, either pre- or post-Meth administration, significantly blocked the DAT reduction. [90]. Administration of minocycline was also effective for Meth-related psychotic disorders as revealed in a clinical case report from Japan [144]. However, only one patient was involved in that clinical study, which was insufficient to perform the statistical analysis. In the latest years, minocycline was found to block the rewarding effect of Meth and reduce the self-administrated amount of Meth on both experimental animal models and human studies [145,146]. Consecutive administration of minocycline (20-40 mg/kg) ameliorated the Meth-induced impairment of long-term memory [147]. Most importantly, minocycline attenuated the maintenance and reinstatement of Meth-seeking behaviors, which indicated that minocycline treatment might be able to reduce the craving and relapse for Meth addiction [148]. Taken together, minocycline might break the aforementioned feedback loop to help Meth-dependent individuals quit from relapse. However, we also noticed that there are some divergent views in this regard. In contrast to the researches mentioned above, there are some other studies suggesting that minocycline is unable to protect neurons from Meth challenge [89,149]. The discrepancies may, at least in part, be attributed by different doses of Meth employed in these studies (10 mg/kg and 20 mg/kg). The total amount of Meth administrated with multiple times was much higher in those other studies.
The interaction of the stress and Meth abuse has been well-studied. The unpredictable stress was not only considered as a potentiating factor but also a relapse inducer [16,150]. Ketoprofen, an FDA-proved medication previously used to treat arthritis has recently been found to have a potential therapeutic effect on stress-induced inflammatory response in Meth-administrated rats [151,152]. Moreover, persistent stress was also linked with increased microglial activation [153,154]. To investigate the neuroinflammatory effects on stress-induced potentiation of Meth toxicity, the researchers examined the activity of cyclooxygenase (COX), a well-known neuroinflammatory mediator, in rats exposed to both stress and Meth. Their results showed that the COX inhibitor, ketoprofen, attenuated the enhanced monoaminergic toxicity induced by stress as well as Meth administration [155]. Further experiments demonstrated that an increased permeability of BBB induced by neuroinflammation might underlie the synergistic mechanism of stress and Meth. Ketoprofen, applied either during or post the treatment, significantly reduced the impairment of BBB [151]. Despite its therapeutic potential on synergistic effects of Meth and stress, ketoprofen, however, did not have a protective effect on Meth alone. This is consistent with a previous report regarding to the development of COX as a potential intervention target on Meth toxicity [156]. The current results also suggest that neuroinflammation may exacerbate the existing monoaminergic damage, and thus, promote the disease progression [157].
Sigma receptor (Sig-R), an endoplasmic reticulum protein, has two subtypes expressed in the brain (Sig-1R and Sig-2R) [158]. Meth binds to Sig-R with preferential binding affinity on Sig-1R (2-4 μM), ten times higher than Sig-2R (16-47 μM) [159]. Sig-1R has been implicated in the addiction and toxicity induced by Meth [160-164]. The adaptive upregulation of Sig-1R in the mid-brain was observed after 5 weeks of self-administrated with Meth [165]. Antagonists of Sig-1R block the Meth-induced neurotoxic effects [163] and prevent the development of behavioral sensitization to Meth [164]. In the CNS, Sig-1R is expressed in microglia [166]. A sigma receptor antagonist (SN79) was found to suppress microglial activation and proinflammatory cytokine release in vivo, which blocks the subsequent Meth-induced neurotoxicity. In vitro, inhibition of Sig-1R blocked the Meth-induced microglial apoptosis [167] and with pretreatment of Ditolylguanidine (DTG) or afobazole, sigma receptor agonists, the ATP-induced Ca2+ in microglia decreased and proinflammatory cytokine expression reduced [168]. Although these results suggest sigma receptor as a viable target, it is still too early to conclude that blocking Sig-1R reverse the Meth-induced toxicity by modulation of microglial activity. First, localization studies indicated that Sig-1R was not exclusively expressed in microglia [169]. Second, SN79 is also reported with attenuation of Meth-induced astrogliosis [170]. In cultured astrocyte, Sig-1R is involved in Meth-induced astrocyte activation in a positive feedback manner [171]. More studies focused on the exact biochemical relationship between Sig-1R and Meth in microglia are needed. The interaction of Sig-1R and inositol 1,4,5-triphosphate receptors (IP3Rs) on mitochondria-associated ER membrane (MAM) is worth to be investigated on cultured microglia treated with Meth. The potentiation of Ca2+ transmission between ER and mitochondria might play a role in Meth-induced microglial activation [172].
The most striking progress having been made recent years based on neuroinflammation is the use of ibudilast to treat Meth dependence. Ibudilast exerts its anti-inflammation effect by inhibition of phsphodiesterase-4 [173,174]. Because its ability to cross the BBB and suppress the microglial activation, ibudilast has been thoroughly studied for potential efficacy on Meth addiction. To date, Ibudilast has been demonstrated to be effective in reducing the self-administration of Meth and rate of stress-induced Meth relapse [16,146]. Moreover, it is also showed to modulate Meth-induced behavioral change [15]. The neuroprotective role of this drug has been attributed to its inhibition of inflammatory response [175]. Given the fact that Meth is prevalent in HIV-infected patients and microglia are one the most important common targets that mediate the rapid progression of HAND in HIV patients with Meth abuse, it is very meaningful that ibudilast also inhibits Tat-induced proinflammatory cytokines release from microglia [176]. Altogether, this pharmacotherapy is quite promising for Meth addiction and may block the potentiation effect of Meth on the pre-existing neurodegenerative disorders, such as HAND. Currently, using ibudilast for Meth dependence has completed the phase I clinical trial for drug safety, and now is undertaking the phase II clinical trial (NCT01860807).
Summary
Traditionally, inflammation has four common cellular and molecular hallmarks: upregulation of proinflammatory cytokines and chemokines, activation macrophages ( and brain microglia), recruitment of leukocytes and tissue damage [177]. In the study of Meth-induced neuroinflammation, solid evidence demonstrate that administration of Meth could induce a substantial inflammatory response [33] as shown in Figure 1. Upregulated inflammatory mediators and proliferation of microglia were found highly correlated with subsequent Meth-induced neurotoxicity [18,81]. The Meth-induced microglial activation temporally precedes the neuronal damage and response to Meth application in a dose-dependent manner, suggesting a specific causal relationship with Meth-induced neurotoxicity [18,178]. In human abusers, overactivated microglia have been found in multiple brain areas, which exist for years even in Meth abstainers [19]. Taking together, these results indicate neuroinflammation is a potential target for Meth-induced neurotoxicity. Multiple pharmacotherapies targeting neuroinflammation have been carried out in experimental animals and demonstrated neuroprotective effects against the Meth-induced neurotoxicity (Figure 1). However, because of the limited information on Meth’s target receptors in microglia, the precise molecular mechanism(s) for these protective drugs are still not fully understood. Given the possibility that the inflammatory responses of microglia after Meth stimulation in vivo are a secondary response to the neuronal DAMPs (i.e., HMGB1 or Dopamine-quoin), it is worth to investigate whether Meth could interact with other neuronal toxic factors to enhance the microglial activation in a synergistic manner. All in all, identification of Meth specific molecular receptor(s) in microglia will allow us to develop more specific therapeutic strategies for not only eventually helping abusers quit from Meth, but also preventing or ameliorating Meth-induced neurotoxicity.
Disclosure of conflict of interest
None.
References
- 1.Degenhardt L, Mathers B, Guarinieri M, Panda S, Phillips B, Strathdee SA, Tyndall M, Wiessing L, Wodak A, Howard J Reference Group to the United Nations on HIV and injecting drug use. Meth/amphetamine use and associated HIV: Implications for global policy and public health. Int J Drug Policy. 2010;21:347–358. doi: 10.1016/j.drugpo.2009.11.007. [DOI] [PubMed] [Google Scholar]
- 2.Volkow ND, Chang L, Wang GJ, Fowler JS, Leonido-Yee M, Franceschi D, Sedler MJ, Gatley SJ, Hitzemann R, Ding YS, Logan J, Wong C, Miller EN. Association of dopamine transporter reduction with psychomotor impairment in methamphetamine abusers. Am J Psychiatry. 2001;158:377–382. doi: 10.1176/appi.ajp.158.3.377. [DOI] [PubMed] [Google Scholar]
- 3.Volkow ND, Li TK. Drug addiction: the neurobiology of behaviour gone awry. Nat Rev Neurosci. 2004;5:963–970. doi: 10.1038/nrn1539. [DOI] [PubMed] [Google Scholar]
- 4.Sekine Y, Ouchi Y, Takei N, Yoshikawa E, Nakamura K, Futatsubashi M, Okada H, Minabe Y, Suzuki K, Iwata Y, Tsuchiya KJ, Tsukada H, Iyo M, Mori N. Brain serotonin transporter density and aggression in abstinent methamphetamine abusers. Arch Gen Psychiatry. 2006;63:90–100. doi: 10.1001/archpsyc.63.1.90. [DOI] [PubMed] [Google Scholar]
- 5.Goldstein RZ, Volkow ND. Dysfunction of the prefrontal cortex in addiction: neuroimaging findings and clinical implications. Nat Rev Neurosci. 2011;12:652–669. doi: 10.1038/nrn3119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Panenka WJ, Procyshyn RM, Lecomte T, MacEwan GW, Flynn SW, Honer WG, Barr AM. Methamphetamine use: a comprehensive review of molecular, preclinical and clinical findings. Drug Alcohol Depend. 2013;129:167–179. doi: 10.1016/j.drugalcdep.2012.11.016. [DOI] [PubMed] [Google Scholar]
- 7.Melega WP, Lacan G, Harvey DC, Huang SC, Phelps ME. Dizocilpine and reduced body temperature do not prevent methamphetamine-induced neurotoxicity in the vervet monkey: [11C] WIN 35,428-positron emission tomography studies. Neurosci Lett. 1998;258:17–20. doi: 10.1016/s0304-3940(98)00845-3. [DOI] [PubMed] [Google Scholar]
- 8.Villemagne V, Yuan J, Wong DF, Dannals RF, Hatzidimitriou G, Mathews WB, Ravert HT, Musachio J, McCann UD, Ricaurte GA. Brain dopamine neurotoxicity in baboons treated with doses of methamphetamine comparable to those recreationally abused by humans: evidence from [11C] WIN-35,428 positron emission tomography studies and direct in vitro determinations. J Neurosci. 1998;18:419–427. doi: 10.1523/JNEUROSCI.18-01-00419.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Thompson PM, Hayashi KM, Simon SL, Geaga JA, Hong MS, Sui Y, Lee JY, Toga AW, Ling W, London ED. Structural abnormalities in the brains of human subjects who use methamphetamine. J Neurosci. 2004;24:6028–6036. doi: 10.1523/JNEUROSCI.0713-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Marshall JF, Belcher AM, Feinstein EM, O’Dell SJ. Methamphetamine-induced neural and cognitive changes in rodents. Addiction. 2007;102(Suppl 1):61–69. doi: 10.1111/j.1360-0443.2006.01780.x. [DOI] [PubMed] [Google Scholar]
- 11.O’Dell SJ, Feinberg LM, Marshall JF. A neurotoxic regimen of methamphetamine impairs novelty recognition as measured by a social odor-based task. Behav Brain Res. 2011;216:396–401. doi: 10.1016/j.bbr.2010.08.022. [DOI] [PubMed] [Google Scholar]
- 12.Schroder N, O’Dell SJ, Marshall JF. Neurotoxic methamphetamine regimen severely impairs recognition memory in rats. Synapse. 2003;49:89–96. doi: 10.1002/syn.10210. [DOI] [PubMed] [Google Scholar]
- 13.Kalechstein AD, Newton TF, Green M. Methamphetamine dependence is associated with neurocognitive impairment in the initial phases of abstinence. J Neuropsychiatry Clin Neurosci. 2003;15:215–220. doi: 10.1176/jnp.15.2.215. [DOI] [PubMed] [Google Scholar]
- 14.Worley MJ, Heinzerling KG, Roche DJ, Shoptaw S. Ibudilast attenuates subjective effects of methamphetamine in a placebo-controlled inpatient study. Drug Alcohol Depend. 2016;162:245–250. doi: 10.1016/j.drugalcdep.2016.02.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Snider SE, Vunck SA, van den Oord EJ, Adkins DE, McClay JL, Beardsley PM. The glial cell modulators, ibudilast and its amino analog, AV1013, attenuate methamphetamine locomotor activity and its sensitization in mice. Eur J Pharmacol. 2012;679:75–80. doi: 10.1016/j.ejphar.2012.01.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Beardsley PM, Shelton KL, Hendrick E, Johnson KW. The glial cell modulator and phosphodiesterase inhibitor, AV411 (ibudilast), attenuates prime- and stress-induced methamphetamine relapse. Eur J Pharmacol. 2010;637:102–108. doi: 10.1016/j.ejphar.2010.04.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Charntikov S, Pittenger ST, Thapa I, Bastola DR, Bevins RA, Pendyala G. Ibudilast reverses the decrease in the synaptic signaling protein phosphatidylethanolamine-binding protein 1 (PEBP1) produced by chronic methamphetamine intake in rats. Drug Alcohol Depend. 2015;152:15–23. doi: 10.1016/j.drugalcdep.2015.04.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.LaVoie MJ, Card JP, Hastings TG. Microglial activation precedes dopamine terminal pathology in methamphetamine-induced neurotoxicity. Exp Neurol. 2004;187:47–57. doi: 10.1016/j.expneurol.2004.01.010. [DOI] [PubMed] [Google Scholar]
- 19.Sekine Y, Ouchi Y, Sugihara G, Takei N, Yoshikawa E, Nakamura K, Iwata Y, Tsuchiya KJ, Suda S, Suzuki K, Kawai M, Takebayashi K, Yamamoto S, Matsuzaki H, Ueki T, Mori N, Gold MS, Cadet JL. Methamphetamine causes microglial activation in the brains of human abusers. J Neurosci. 2008;28:5756–5761. doi: 10.1523/JNEUROSCI.1179-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Narendran R, Lopresti BJ, Mason NS, Deuitch L, Paris J, Himes ML, Kodavali CV, Nimgaonkar VL. Cocaine abuse in humans is not associated with increased microglial activation: an 18-kDa translocator protein positron emission tomography imaging study with [11C] PBR28. J Neurosci. 2014;34:9945–9950. doi: 10.1523/JNEUROSCI.0928-14.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Thomas DM, Walker PD, Benjamins JA, Geddes TJ, Kuhn DM. Methamphetamine neurotoxicity in dopamine nerve endings of the striatum is associated with microglial activation. J Pharmacol Exp Ther. 2004;311:1–7. doi: 10.1124/jpet.104.070961. [DOI] [PubMed] [Google Scholar]
- 22.Biber K, Neumann H, Inoue K, Boddeke HW. Neuronal ‘On’ and ‘Off’ signals control microglia. Trends Neurosci. 2007;30:596–602. doi: 10.1016/j.tins.2007.08.007. [DOI] [PubMed] [Google Scholar]
- 23.Hellwig S, Heinrich A, Biber K. The brain’s best friend: microglial neurotoxicity revisited. Front Cell Neurosci. 2013;7:71. doi: 10.3389/fncel.2013.00071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Prinz M, Mildner A. Microglia in the CNS: immigrants from another world. Glia. 2011;59:177–187. doi: 10.1002/glia.21104. [DOI] [PubMed] [Google Scholar]
- 25.Thomas DM, Francescutti-Verbeem DM, Kuhn DM. Methamphetamine-induced neurotoxicity and microglial activation are not mediated by fractalkine receptor signaling. J Neurochem. 2008;106:696–705. doi: 10.1111/j.1471-4159.2008.05421.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Yue X, Qiao D, Wang A, Tan X, Li Y, Liu C, Wang H. CD200 attenuates methamphetamine-induced microglial activation and dopamine depletion. J Huazhong Univ Sci Technolog Med Sci. 2012;32:415–421. doi: 10.1007/s11596-012-0072-0. [DOI] [PubMed] [Google Scholar]
- 27.Block ML, Zecca L, Hong JS. Microglia-mediated neurotoxicity: uncovering the molecular mechanisms. Nat Rev Neurosci. 2007;8:57–69. doi: 10.1038/nrn2038. [DOI] [PubMed] [Google Scholar]
- 28.Loftis JM, Choi D, Hoffman W, Huckans MS. Methamphetamine causes persistent immune dysregulation: a cross-species, translational report. Neurotox Res. 2011;20:59–68. doi: 10.1007/s12640-010-9223-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Mata MM, Napier TC, Graves SM, Mahmood F, Raeisi S, Baum LL. Methamphetamine decreases CD4 T cell frequency and alters proinflammatory cytokine production in a model of drug abuse. Eur J Pharmacol. 2015;752:26–33. doi: 10.1016/j.ejphar.2015.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Harms R, Morsey B, Boyer CW, Fox HS, Sarvetnick N. Methamphetamine administration targets multiple immune subsets and induces phenotypic alterations suggestive of immunosuppression. PLoS One. 2012;7:e49897. doi: 10.1371/journal.pone.0049897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.In SW, Son EW, Rhee DK, Pyo S. Modulation of murine macrophage function by methamphetamine. J Toxicol Environ Health A. 2004;67:1923–1937. doi: 10.1080/15287390490514589. [DOI] [PubMed] [Google Scholar]
- 32.Nair MP, Mahajan S, Sykes D, Bapardekar MV, Reynolds JL. Methamphetamine modulates DC-SIGN expression by mature dendritic cells. J Neuroimmune Pharmacol. 2006;1:296–304. doi: 10.1007/s11481-006-9027-1. [DOI] [PubMed] [Google Scholar]
- 33.McConnell SE, O’Banion MK, Cory-Slechta DA, Olschowka JA, Opanashuk LA. Characterization of binge-dosed methamphetamine-induced neurotoxicity and neuroinflammation. Neurotoxicology. 2015;50:131–141. doi: 10.1016/j.neuro.2015.08.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Najera JA, Bustamante EA, Bortell N, Morsey B, Fox HS, Ravasi T, Marcondes MC. Methamphetamine abuse affects gene expression in brain-derived microglia of SIV-infected macaques to enhance inflammation and promote virus targets. BMC Immunol. 2016;17:7. doi: 10.1186/s12865-016-0145-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Downey LA, Loftis JM. Altered energy production, lowered antioxidant potential, and inflammatory processes mediate CNS damage associated with abuse of the psychostimulants MDMA and methamphetamine. Eur J Pharmacol. 2014;727:125–129. doi: 10.1016/j.ejphar.2014.01.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Frank MG, Adhikary S, Sobesky JL, Weber MD, Watkins LR, Maier SF. The danger-associated molecular pattern HMGB1 mediates the neuroinflammatory effects of methamphetamine. Brain Behav Immun. 2016;51:99–108. doi: 10.1016/j.bbi.2015.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Colfax G, Shoptaw S. The methamphetamine epidemic: implications for HIV prevention and treatment. Curr HIV/AIDS Rep. 2005;2:194–199. doi: 10.1007/s11904-005-0016-4. [DOI] [PubMed] [Google Scholar]
- 38.Jernigan TL, Gamst AC, Archibald SL, Fennema-Notestine C, Mindt MR, Marcotte TD, Heaton RK, Ellis RJ, Grant I. Effects of methamphetamine dependence and HIV infection on cerebral morphology. Am J Psychiatry. 2005;162:1461–1472. doi: 10.1176/appi.ajp.162.8.1461. [DOI] [PubMed] [Google Scholar]
- 39.Kesby JP, Heaton RK, Young JW, Umlauf A, Woods SP, Letendre SL, Markou A, Grant I, Semenova S. Methamphetamine exposure combined with HIV-1 disease or gp120 expression: comparison of learning and executive functions in humans and mice. Neuropsychopharmacology. 2015;40:1899–1909. doi: 10.1038/npp.2015.39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Kesby JP, Markou A, Semenova S Translational Methamphetamine AIDS Research Center (TMARC) Group. Cognitive deficits associated with combined HIV gp120 expression and chronic methamphetamine exposure in mice. Eur Neuropsychopharmacol. 2015;25:141–150. doi: 10.1016/j.euroneuro.2014.07.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Hoefer MM, Sanchez AB, Maung R, de Rozieres CM, Catalan IC, Dowling CC, Thaney VE, Pina-Crespo J, Zhang D, Roberts AJ, Kaul M. Combination of methamphetamine and HIV-1 gp120 causes distinct long-term alterations of behavior, gene expression, and injury in the central nervous system. Exp Neurol. 2015;263:221–234. doi: 10.1016/j.expneurol.2014.09.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Henry BL, Geyer MA, Buell M, Perry W, Young JW, Minassian A Translational Methamphetamine AIDS Research Center (TMARC) Group. Behavioral effects of chronic methamphetamine treatment in HIV-1 gp120 transgenic mice. Behav Brain Res. 2013;236:210–220. doi: 10.1016/j.bbr.2012.08.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Roberts AJ, Maung R, Sejbuk NE, Ake C, Kaul M. Alteration of Methamphetamine-induced stereotypic behaviour in transgenic mice expressing HIV-1 envelope protein gp120. J Neurosci Methods. 2010;186:222–225. doi: 10.1016/j.jneumeth.2009.11.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Langford D, Adame A, Grigorian A, Grant I, McCutchan JA, Ellis RJ, Marcotte TD, Masliah E HIV Neurobehavioral Research Center Group. Patterns of selective neuronal damage in methamphetamine-user AIDS patients. J Acquir Immune Defic Syndr. 2003;34:467–474. doi: 10.1097/00126334-200312150-00004. [DOI] [PubMed] [Google Scholar]
- 45.Patel D, Desai GM, Frases S, Cordero RJ, DeLeon-Rodriguez CM, Eugenin EA, Nosanchuk JD, Martinez LR. Methamphetamine enhances Cryptococcus neoformans pulmonary infection and dissemination to the brain. MBio. 2013:4. doi: 10.1128/mBio.00400-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Eugenin EA, Greco JM, Frases S, Nosanchuk JD, Martinez LR. Methamphetamine alters blood brain barrier protein expression in mice, facilitating central nervous system infection by neurotropic Cryptococcus neoformans. J Infect Dis. 2013;208:699–704. doi: 10.1093/infdis/jit117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Liu X, Silverstein PS, Singh V, Shah A, Qureshi N, Kumar A. Methamphetamine increases LPS-mediated expression of IL-8, TNF-alpha and IL-1beta in human macrophages through common signaling pathways. PLoS One. 2012;7:e33822. doi: 10.1371/journal.pone.0033822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Jung BD, Shin EJ, Nguyen XK, Jin CH, Bach JH, Park SJ, Nah SY, Wie MB, Bing G, Kim HC. Potentiation of methamphetamine neurotoxicity by intrastriatal lipopolysaccharide administration. Neurochem Int. 2010;56:229–244. doi: 10.1016/j.neuint.2009.10.005. [DOI] [PubMed] [Google Scholar]
- 49.Cruickshank CC, Dyer KR. A review of the clinical pharmacology of methamphetamine. Addiction. 2009;104:1085–1099. doi: 10.1111/j.1360-0443.2009.02564.x. [DOI] [PubMed] [Google Scholar]
- 50.Kish SJ. Pharmacologic mechanisms of crystal meth. CMAJ. 2008;178:1679–1682. doi: 10.1503/cmaj.071675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Sulzer D, Sonders MS, Poulsen NW, Galli A. Mechanisms of neurotransmitter release by amphetamines: a review. Prog Neurobiol. 2005;75:406–433. doi: 10.1016/j.pneurobio.2005.04.003. [DOI] [PubMed] [Google Scholar]
- 52.Brown JM, Hanson GR, Fleckenstein AE. Methamphetamine rapidly decreases vesicular dopamine uptake. J Neurochem. 2000;74:2221–2223. doi: 10.1046/j.1471-4159.2000.0742221.x. [DOI] [PubMed] [Google Scholar]
- 53.Fleckenstein AE, Metzger RR, Wilkins DG, Gibb JW, Hanson GR. Rapid and reversible effects of methamphetamine on dopamine transporters. J Pharmacol Exp Ther. 1997;282:834–838. [PubMed] [Google Scholar]
- 54.Kuczenski R, Segal DS, Aizenstein ML. Amphetamine, cocaine, and fencamfamine: relationship between locomotor and stereotypy response profiles and caudate and accumbens dopamine dynamics. J Neurosci. 1991;11:2703–2712. doi: 10.1523/JNEUROSCI.11-09-02703.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Darke S, Kaye S, McKetin R, Duflou J. Major physical and psychological harms of methamphetamine use. Drug Alcohol Rev. 2008;27:253–262. doi: 10.1080/09595230801923702. [DOI] [PubMed] [Google Scholar]
- 56.Simon SL, Domier CP, Sim T, Richardson K, Rawson RA, Ling W. Cognitive performance of current methamphetamine and cocaine abusers. J Addict Dis. 2002;21:61–74. doi: 10.1300/j069v21n01_06. [DOI] [PubMed] [Google Scholar]
- 57.Johanson CE, Frey KA, Lundahl LH, Keenan P, Lockhart N, Roll J, Galloway GP, Koeppe RA, Kilbourn MR, Robbins T, Schuster CR. Cognitive function and nigrostriatal markers in abstinent methamphetamine abusers. Psychopharmacology (Berl) 2006;185:327–338. doi: 10.1007/s00213-006-0330-6. [DOI] [PubMed] [Google Scholar]
- 58.Volkow ND. Drug abuse and mental illness: progress in understanding comorbidity. Am J Psychiatry. 2001;158:1181–1183. doi: 10.1176/appi.ajp.158.8.1181. [DOI] [PubMed] [Google Scholar]
- 59.Frey K, Kilbourn M, Robinson T. Reduced striatal vesicular monoamine transporters after neurotoxic but not after behaviorally-sensitizing doses of methamphetamine. Eur J Pharmacol. 1997;334:273–279. doi: 10.1016/s0014-2999(97)01152-7. [DOI] [PubMed] [Google Scholar]
- 60.Sekine Y, Iyo M, Ouchi Y, Matsunaga T, Tsukada H, Okada H, Yoshikawa E, Futatsubashi M, Takei N, Mori N. Methamphetamine-related psychiatric symptoms and reduced brain dopamine transporters studied with PET. Am J Psychiatry. 2001;158:1206–1214. doi: 10.1176/appi.ajp.158.8.1206. [DOI] [PubMed] [Google Scholar]
- 61.Bae SC, Lyoo IK, Sung YH, Yoo J, Chung A, Yoon SJ, Kim DJ, Hwang J, Kim SJ, Renshaw PF. Increased white matter hyperintensities in male methamphetamine abusers. Drug Alcohol Depend. 2006;81:83–88. doi: 10.1016/j.drugalcdep.2005.05.016. [DOI] [PubMed] [Google Scholar]
- 62.Kettenmann H, Hanisch UK, Noda M, Verkhratsky A. Physiology of microglia. Physiol Rev. 2011;91:461–553. doi: 10.1152/physrev.00011.2010. [DOI] [PubMed] [Google Scholar]
- 63.Lull ME, Block ML. Microglial activation and chronic neurodegeneration. Neurotherapeutics. 2010;7:354–365. doi: 10.1016/j.nurt.2010.05.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Escubedo E, Guitart L, Sureda FX, Jimenez A, Pubill D, Pallas M, Camins A, Camarasa J. Microgliosis and down-regulation of adenosine transporter induced by methamphetamine in rats. Brain Res. 1998;814:120–126. doi: 10.1016/s0006-8993(98)01065-8. [DOI] [PubMed] [Google Scholar]
- 65.Bowyer JF, Ali S. High doses of methamphetamine that cause disruption of the bloodbrain barrier in limbic regions produce extensive neuronal degeneration in mouse hippocampus. Synapse. 2006;60:521–532. doi: 10.1002/syn.20324. [DOI] [PubMed] [Google Scholar]
- 66.Quinton MS, Yamamoto BK. Causes and consequences of methamphetamine and MDMA toxicity. AAPS J. 2006;8:E337–347. doi: 10.1007/BF02854904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Ramirez SH, Potula R, Fan S, Eidem T, Papugani A, Reichenbach N, Dykstra H, Weksler BB, Romero IA, Couraud PO, Persidsky Y. Methamphetamine disrupts blood-brain barrier function by induction of oxidative stress in brain endothelial cells. J Cereb Blood Flow Metab. 2009;29:1933–1945. doi: 10.1038/jcbfm.2009.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Park M, Kim HJ, Lim B, Wylegala A, Toborek M. Methamphetamine-induced occludin endocytosis is mediated by the Arp2/3 complexregulated actin rearrangement. J Biol Chem. 2013;288:33324–33334. doi: 10.1074/jbc.M113.483487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Bennett BA, Hyde CE, Pecora JR, Clodfelter JE. Long-term cocaine administration is not neurotoxic to cultured fetal mesencephalic dopamine neurons. Neurosci Lett. 1993;153:210–214. doi: 10.1016/0304-3940(93)90324-e. [DOI] [PubMed] [Google Scholar]
- 70.O’Callaghan JP, Miller DB. Neurotoxicity profiles of substituted amphetamines in the C57BL/6J mouse. J Pharmacol Exp Ther. 1994;270:741–751. [PubMed] [Google Scholar]
- 71.Thomas DM, Kuhn DM. MK-801 and dextromethorphan block microglial activation and protect against methamphetamine-induced neurotoxicity. Brain Res. 2005;1050:190–198. doi: 10.1016/j.brainres.2005.05.049. [DOI] [PubMed] [Google Scholar]
- 72.Gygi MP, Gygi SP, Johnson M, Wilkins DG, Gibb JW, Hanson GR. Mechanisms for tolerance to methamphetamine effects. Neuropharmacology. 1996;35:751–757. doi: 10.1016/0028-3908(96)84647-8. [DOI] [PubMed] [Google Scholar]
- 73.Stephans S, Yamamoto B. Methamphetamines pretreatment and the vulnerability of the striatum to methamphetamine neurotoxicity. Neuroscience. 1996;72:593–600. doi: 10.1016/0306-4522(95)00587-0. [DOI] [PubMed] [Google Scholar]
- 74.Riddle EL, Kokoshka JM, Wilkins DG, Hanson GR, Fleckenstein AE. Tolerance to the neurotoxic effects of methamphetamine in young rats. Eur J Pharmacol. 2002;435:181–185. doi: 10.1016/s0014-2999(01)01592-8. [DOI] [PubMed] [Google Scholar]
- 75.Thomas DM, Kuhn DM. Attenuated microglial activation mediates tolerance to the neurotoxic effects of methamphetamine. J Neurochem. 2005;92:790–797. doi: 10.1111/j.1471-4159.2004.02906.x. [DOI] [PubMed] [Google Scholar]
- 76.Friend DM, Keefe KA. Glial reactivity in resistance to methamphetamine-induced neurotoxicity. J Neurochem. 2013;125:566–574. doi: 10.1111/jnc.12201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Buchanan JB, Sparkman NL, Johnson RW. A neurotoxic regimen of methamphetamine exacerbates the febrile and neuroinflammatory response to a subsequent peripheral immune stimulus. J Neuroinflammation. 2010;7:82. doi: 10.1186/1742-2094-7-82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Clark KH, Wiley CA, Bradberry CW. Psychostimulant abuse and neuroinflammation: emerging evidence of their interconnection. Neurotox Res. 2013;23:174–188. doi: 10.1007/s12640-012-9334-7. [DOI] [PubMed] [Google Scholar]
- 79.Coelho-Santos V, Goncalves J, Fontes-Ribeiro C, Silva AP. Prevention of methamphetamineinduced microglial cell death by TNF-alpha and IL-6 through activation of the JAK-STAT pathway. J Neuroinflammation. 2012;9:103. doi: 10.1186/1742-2094-9-103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Wires ES, Alvarez D, Dobrowolski C, Wang Y, Morales M, Karn J, Harvey BK. Methamphetamine activates nuclear factor kappalight-chain-enhancer of activated B cells (NFkappaB) and induces human immunodeficiency virus (HIV) transcription in human microglial cells. J Neurovirol. 2012;18:400–410. doi: 10.1007/s13365-012-0103-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Kuhn DM, Francescutti-Verbeem DM, Thomas DM. Dopamine quinones activate microglia and induce a neurotoxic gene expression profile: relationship to methamphetamine-induced nerve ending damage. Ann N Y Acad Sci. 2006;1074:31–41. doi: 10.1196/annals.1369.003. [DOI] [PubMed] [Google Scholar]
- 82.Miyazaki I, Asanuma M, Diaz-Corrales FJ, Fukuda M, Kitaichi K, Miyoshi K, Ogawa N. Methamphetamine-induced dopaminergic neurotoxicity is regulated by quinone-formation-related molecules. FASEB J. 2006;20:571–573. doi: 10.1096/fj.05-4996fje. [DOI] [PubMed] [Google Scholar]
- 83.Thomas DM, Francescutti-Verbeem DM, Kuhn DM. The newly synthesized pool of dopamine determines the severity of methamphetamine-induced neurotoxicity. J Neurochem. 2008;105:605–616. doi: 10.1111/j.1471-4159.2007.05155.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Chao CC, Hu S, Molitor TW, Shaskan EG, Peterson PK. Activated microglia mediate neuronal cell injury via a nitric oxide mechanism. J Immunol. 1992;149:2736–2741. [PubMed] [Google Scholar]
- 85.Gao HM, Zhou H, Zhang F, Wilson BC, Kam W, Hong JS. HMGB1 acts on microglia Mac1 to mediate chronic neuroinflammation that drives progressive neurodegeneration. J Neurosci. 2011;31:1081–1092. doi: 10.1523/JNEUROSCI.3732-10.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Lehnardt S, Schott E, Trimbuch T, Laubisch D, Krueger C, Wulczyn G, Nitsch R, Weber JR. A vicious cycle involving release of heat shock protein 60 from injured cells and activation of toll-like receptor 4 mediates neurodegeneration in the CNS. J Neurosci. 2008;28:2320–2331. doi: 10.1523/JNEUROSCI.4760-07.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Levesque S, Wilson B, Gregoria V, Thorpe LB, Dallas S, Polikov VS, Hong JS, Block ML. Reactive microgliosis: extracellular micro-calpain and microglia-mediated dopaminergic neurotoxicity. Brain. 2010;133:808–821. doi: 10.1093/brain/awp333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Pais TF, Figueiredo C, Peixoto R, Braz MH, Chatterjee S. Necrotic neurons enhance microglial neurotoxicity through induction of glutaminase by a MyD88-dependent pathway. J Neuroinflammation. 2008;5:43. doi: 10.1186/1742-2094-5-43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Sriram K, Miller DB, O’Callaghan JP. Minocycline attenuates microglial activation but fails to mitigate striatal dopaminergic neurotoxicity: role of tumor necrosis factor-alpha. J Neurochem. 2006;96:706–718. doi: 10.1111/j.1471-4159.2005.03566.x. [DOI] [PubMed] [Google Scholar]
- 90.Hashimoto K, Tsukada H, Nishiyama S, Fukumoto D, Kakiuchi T, Iyo M. Protective effects of minocycline on the reduction of dopamine transporters in the striatum after administration of methamphetamine: a positron emission tomography study in conscious monkeys. Biol Psychiatry. 2007;61:577–581. doi: 10.1016/j.biopsych.2006.03.019. [DOI] [PubMed] [Google Scholar]
- 91.Kelly KA, Miller DB, Bowyer JF, O’Callaghan JP. Chronic exposure to corticosterone enhances the neuroinflammatory and neurotoxic responses to methamphetamine. J Neurochem. 2012;122:995–1009. doi: 10.1111/j.1471-4159.2012.07864.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Zhang X, Dong F, Mayer GE, Bruch DC, Ren J, Culver B. Selective inhibition of cyclooxygenase-2 exacerbates methamphetamine-induced dopamine depletion in the striatum in rats. Neuroscience. 2007;150:950–958. doi: 10.1016/j.neuroscience.2007.09.059. [DOI] [PubMed] [Google Scholar]
- 93.Chen MF, Gill AJ, Kolson DL. Neuropathogenesis of HIV-associated neurocognitive disorders: roles for immune activation, HIV blipping and viral tropism. Curr Opin HIV AIDS. 2014;9:559–564. doi: 10.1097/COH.0000000000000105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Everall I, Vaida F, Khanlou N, Lazzaretto D, Achim C, Letendre S, Moore D, Ellis R, Cherner M, Gelman B, Morgello S, Singer E, Grant I, Masliah E National NeuroAIDS Tissue Consortium (NNTC) Cliniconeuropathologic correlates of human immunodeficiency virus in the era of antiretroviral therapy. J Neurovirol. 2009;15:360–370. doi: 10.3109/13550280903131915. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Kumar AM, Borodowsky I, Fernandez B, Gonzalez L, Kumar M. Human immunodeficiency virus type 1 RNA Levels in different regions of human brain: quantification using real-time reverse transcriptase-polymerase chain reaction. J Neurovirol. 2007;13:210–224. doi: 10.1080/13550280701327038. [DOI] [PubMed] [Google Scholar]
- 96.Harezlak J, Buchthal S, Taylor M, Schifitto G, Zhong J, Daar E, Alger J, Singer E, Campbell T, Yiannoutsos C, Cohen R, Navia B HIV Neuroimaging Consortium. Persistence of HIV-associated cognitive impairment, inflammation, and neuronal injury in era of highly active antiretroviral treatment. AIDS. 2011;25:625–633. doi: 10.1097/QAD.0b013e3283427da7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Eden A, Price RW, Spudich S, Fuchs D, Hagberg L, Gisslen M. Immune activation of the central nervous system is still present after >4 years of effective highly active antiretroviral therapy. J Infect Dis. 2007;196:1779–1783. doi: 10.1086/523648. [DOI] [PubMed] [Google Scholar]
- 98.Garvey LJ, Pavese N, Politis M, Ramlackhansingh A, Brooks DJ, Taylor-Robinson SD, Winston A. Increased microglia activation in neurologically asymptomatic HIV-infected patients receiving effective ART. AIDS. 2014;28:67–72. doi: 10.1097/01.aids.0000432467.54003.f7. [DOI] [PubMed] [Google Scholar]
- 99.Halkitis PN, Parsons JT, Stirratt MJ. A double epidemic: crystal methamphetamine drug use in relation to HIV transmission among gay men. J Homosex. 2001;41:17–35. doi: 10.1300/J082v41n02_02. [DOI] [PubMed] [Google Scholar]
- 100.Letendre SL, Cherner M, Ellis RJ, Marquie-Beck J, Gragg B, Marcotte T, Heaton RK, McCutchan JA, Grant I HNRC Group. The effects of hepatitis C, HIV, and methamphetamine dependence on neuropsychological performance: biological correlates of disease. AIDS. 2005;19(Suppl 3):S72–78. doi: 10.1097/01.aids.0000192073.18691.ff. [DOI] [PubMed] [Google Scholar]
- 101.Gonzalez-Scarano F, Martin-Garcia J. The neuropathogenesis of AIDS. Nat Rev Immunol. 2005;5:69–81. doi: 10.1038/nri1527. [DOI] [PubMed] [Google Scholar]
- 102.Ellis RJ, Childers ME, Cherner M, Lazzaretto D, Letendre S, Grant I HIV Neurobehavioral Research Center Group. Increased human immunodeficiency virus loads in active methamphetamine users are explained by reduced effectiveness of antiretroviral therapy. J Infect Dis. 2003;188:1820–1826. doi: 10.1086/379894. [DOI] [PubMed] [Google Scholar]
- 103.Carrico AW, Johnson MO, Colfax GN, Moskowitz JT. Affective correlates of stimulant use and adherence to anti-retroviral therapy among HIV-positive methamphetamine users. AIDS Behav. 2010;14:769–777. doi: 10.1007/s10461-008-9513-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Turowski P, Kenny BA. The blood-brain barrier and methamphetamine: open sesame? Front Neurosci. 2015;9:156. doi: 10.3389/fnins.2015.00156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Liang H, Wang X, Chen H, Song L, Ye L, Wang SH, Wang YJ, Zhou L, Ho WZ. Methamphetamine enhances HIV infection of macrophages. Am J Pathol. 2008;172:1617–1624. doi: 10.2353/ajpath.2008.070971. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Nair MP, Saiyed ZM, Nair N, Gandhi NH, Rodriguez JW, Boukli N, Provencio-Vasquez E, Malow RM, Miguez-Burbano MJ. Methamphetamine enhances HIV-1 infectivity in monocyte derived dendritic cells. J Neuroimmune Pharmacol. 2009;4:129–139. doi: 10.1007/s11481-008-9128-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Czub S, Czub M, Koutsilieri E, Sopper S, Villinger F, Muller JG, Stahl-Hennig C, Riederer P, Ter Meulen V, Gosztonyi G. Modulation of simian immunodeficiency virus neuropathology by dopaminergic drugs. Acta Neuropathol. 2004;107:216–226. doi: 10.1007/s00401-003-0801-3. [DOI] [PubMed] [Google Scholar]
- 108.Czub S, Koutsilieri E, Sopper S, Czub M, Stahl-Hennig C, Muller JG, Pedersen V, Gsell W, Heeney JL, Gerlach M, Gosztonyi G, Riederer P, ter Meulen V. Enhancement of central nervous system pathology in early simian immunodeficiency virus infection by dopaminergic drugs. Acta Neuropathol. 2001;101:85–91. doi: 10.1007/s004010000313. [DOI] [PubMed] [Google Scholar]
- 109.Purohit V, Rapaka R, Shurtleff D. Drugs of abuse, dopamine, and HIV-associated neurocognitive disorders/HIV-associated dementia. Mol Neurobiol. 2011;44:102–110. doi: 10.1007/s12035-011-8195-z. [DOI] [PubMed] [Google Scholar]
- 110.Stebbing MJ, Cottee JM, Rana I. The role of ion channels in microglial activation and proliferation-A Complex Interplay between ligandgated ion channels, K(+) channels, and intracellular Ca(2.) Front Immunol. 2015;6:497. doi: 10.3389/fimmu.2015.00497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Liu J, Xu P, Collins C, Liu H, Zhang J, Keblesh JP, Xiong H. HIV-1 Tat protein increases microglial outward K(+) current and resultant neurotoxic activity. PLoS One. 2013;8:e64904. doi: 10.1371/journal.pone.0064904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Xu C, Liu J, Chen L, Liang S, Fujii N, Tamamura H, Xiong H. HIV-1 gp120 enhances outward potassium current via CXCR4 and cAMP-dependent protein kinase A signaling in cultured rat microglia. Glia. 2011;59:997–1007. doi: 10.1002/glia.21171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Wang J, Qian W, Liu J, Zhao J, Yu P, Jiang L, Zhou J, Gao R, Xiao H. Effect of methamphetamine on the microglial damage: role of potassium channel Kv1.3. PLoS One. 2014;9:e88642. doi: 10.1371/journal.pone.0088642. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Goncalves J, Martins T, Ferreira R, Milhazes N, Borges F, Ribeiro CF, Malva JO, Macedo TR, Silva AP. Methamphetamine-induced early increase of IL-6 and TNF-alpha mRNA expression in the mouse brain. Ann N Y Acad Sci. 2008;1139:103–111. doi: 10.1196/annals.1432.043. [DOI] [PubMed] [Google Scholar]
- 115.Nuovo GJ, Alfieri ML. AIDS dementia is associated with massive, activated HIV-1 infection and concomitant expression of several cytokines. Mol Med. 1996;2:358–366. [PMC free article] [PubMed] [Google Scholar]
- 116.Brabers NA, Nottet HS. Role of the pro-inflammatory cytokines TNF-alpha and IL-1beta in HIV-associated dementia. Eur J Clin Invest. 2006;36:447–458. doi: 10.1111/j.1365-2362.2006.01657.x. [DOI] [PubMed] [Google Scholar]
- 117.Lane BR, Lore K, Bock PJ, Andersson J, Coffey MJ, Strieter RM, Markovitz DM. Interleukin-8 stimulates human immunodeficiency virus type 1 replication and is a potential new target for antiretroviral therapy. J Virol. 2001;75:8195–8202. doi: 10.1128/JVI.75.17.8195-8202.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Munoz-Fernandez MA, Navarro J, Garcia A, Punzon C, Fernandez-Cruz E, Fresno M. Replication of human immunodeficiency virus-1 in primary human T cells is dependent on the autocrine secretion of tumor necrosis factor through the control of nuclear factor-kappa B activation. J Allergy Clin Immunol. 1997;100:838–845. doi: 10.1016/s0091-6749(97)70282-3. [DOI] [PubMed] [Google Scholar]
- 119.Fine SM, Angel RA, Perry SW, Epstein LG, Rothstein JD, Dewhurst S, Gelbard HA. Tumor necrosis factor alpha inhibits glutamate uptake by primary human astrocytes. Implications for pathogenesis of HIV-1 dementia. J Biol Chem. 1996;271:15303–15306. doi: 10.1074/jbc.271.26.15303. [DOI] [PubMed] [Google Scholar]
- 120.Mediouni S, Garibaldi Marcondes MC, Miller C, McLaughlin JP, Valente ST. The cross-talk of HIV-1 Tat and methamphetamine in HIV-associated neurocognitive disorders. Front Microbiol. 2015;6:1164. doi: 10.3389/fmicb.2015.01164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Bellinger FP, Madamba S, Siggins GR. Interleukin 1 beta inhibits synaptic strength and long-term potentiation in the rat CA1 hippocampus. Brain Res. 1993;628:227–234. doi: 10.1016/0006-8993(93)90959-q. [DOI] [PubMed] [Google Scholar]
- 122.Xiong H, Boyle J, Winkelbauer M, Gorantla S, Zheng J, Ghorpade A, Persidsky Y, Carlson KA, Gendelman HE. Inhibition of long-term potentiation by interleukin-8: implications for human immunodeficiency virus-1-associated dementia. J Neurosci Res. 2003;71:600–607. doi: 10.1002/jnr.10503. [DOI] [PubMed] [Google Scholar]
- 123.Gage FH, Temple S. Neural stem cells: generating and regenerating the brain. Neuron. 2013;80:588–601. doi: 10.1016/j.neuron.2013.10.037. [DOI] [PubMed] [Google Scholar]
- 124.Pabon MM, Bachstetter AD, Hudson CE, Gemma C, Bickford PC. CX3CL1 reduces neurotoxicity and microglial activation in a rat model of Parkinson’s disease. J Neuroinflammation. 2011;8:9. doi: 10.1186/1742-2094-8-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Cardona AE, Pioro EP, Sasse ME, Kostenko V, Cardona SM, Dijkstra IM, Huang D, Kidd G, Dombrowski S, Dutta R, Lee JC, Cook DN, Jung S, Lira SA, Littman DR, Ransohoff RM. Control of microglial neurotoxicity by the fractalkine receptor. Nat Neurosci. 2006;9:917–924. doi: 10.1038/nn1715. [DOI] [PubMed] [Google Scholar]
- 126.Thomas DM, Francescutti-Verbeem DM, Liu X, Kuhn DM. Identification of differentially regulated transcripts in mouse striatum following methamphetamine treatment--an oligonucleotide microarray approach. J Neurochem. 2004;88:380–393. doi: 10.1046/j.1471-4159.2003.02182.x. [DOI] [PubMed] [Google Scholar]
- 127.Zhang S, Wang XJ, Tian LP, Pan J, Lu GQ, Zhang YJ, Ding JQ, Chen SD. CD200-CD200R dysfunction exacerbates microglial activation and dopaminergic neurodegeneration in a rat model of Parkinson’s disease. J Neuroinflammation. 2011;8:154. doi: 10.1186/1742-2094-8-154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Chitnis T, Imitola J, Wang Y, Elyaman W, Chawla P, Sharuk M, Raddassi K, Bronson RT, Khoury SJ. Elevated neuronal expression of CD200 protects Wlds mice from inflammationmediated neurodegeneration. Am J Pathol. 2007;170:1695–1712. doi: 10.2353/ajpath.2007.060677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Chacko AM, Li C, Pryma DA, Brem S, Coukos G, Muzykantov V. Targeted delivery of antibodybased therapeutic and imaging agents to CNS tumors: crossing the blood-brain barrier divide. Expert Opin Drug Deliv. 2013;10:907–926. doi: 10.1517/17425247.2013.808184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Hillhouse MP, Marinelli-Casey P, Gonzales R, Ang A, Rawson RA Methamphetamine Treatment Project Corporate Authors. Predicting intreatment performance and post-treatment outcomes in methamphetamine users. Addiction. 2007;102(Suppl 1):84–95. doi: 10.1111/j.1360-0443.2007.01768.x. [DOI] [PubMed] [Google Scholar]
- 131.Brecht ML, Herbeck D. Time to relapse following treatment for methamphetamine use: a long-term perspective on patterns and predictors. Drug Alcohol Depend. 2014;139:18–25. doi: 10.1016/j.drugalcdep.2014.02.702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Hoffman WF, Moore M, Templin R, McFarland B, Hitzemann RJ, Mitchell SH. Neuropsychological function and delay discounting in methamphetamine-dependent individuals. Psychopharmacology (Berl) 2006;188:162–170. doi: 10.1007/s00213-006-0494-0. [DOI] [PubMed] [Google Scholar]
- 133.London ED, Simon SL, Berman SM, Mandelkern MA, Lichtman AM, Bramen J, Shinn AK, Miotto K, Learn J, Dong Y, Matochik JA, Kurian V, Newton T, Woods R, Rawson R, Ling W. Mood disturbances and regional cerebral metabolic abnormalities in recently abstinent methamphetamine abusers. Arch Gen Psychiatry. 2004;61:73–84. doi: 10.1001/archpsyc.61.1.73. [DOI] [PubMed] [Google Scholar]
- 134.Berman S, O’Neill J, Fears S, Bartzokis G, London ED. Abuse of amphetamines and structural abnormalities in the brain. Ann N Y Acad Sci. 2008;1141:195–220. doi: 10.1196/annals.1441.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Hoffman WF, Schwartz DL, Huckans MS, McFarland BH, Meiri G, Stevens AA, Mitchell SH. Cortical activation during delay discounting in abstinent methamphetamine dependent individuals. Psychopharmacology (Berl) 2008;201:183–193. doi: 10.1007/s00213-008-1261-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Schwartz DL, Mitchell AD, Lahna DL, Luber HS, Huckans MS, Mitchell SH, Hoffman WF. Global and local morphometric differences in recently abstinent methamphetamine-dependent individuals. Neuroimage. 2010;50:1392–1401. doi: 10.1016/j.neuroimage.2010.01.056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Kim HS, Suh YH. Minocycline and neurodegenerative diseases. Behav Brain Res. 2009;196:168–179. doi: 10.1016/j.bbr.2008.09.040. [DOI] [PubMed] [Google Scholar]
- 138.Beardsley PM, Hauser KF. Glial modulators as potential treatments of psychostimulant abuse. Adv Pharmacol. 2014;69:1–69. doi: 10.1016/B978-0-12-420118-7.00001-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Amin AR, Patel RN, Thakker GD, Lowenstein CJ, Attur MG, Abramson SB. Post-transcriptional regulation of inducible nitric oxide synthase mRNA in murine macrophages by doxycycline and chemically modified tetracyclines. FEBS Lett. 1997;410:259–264. doi: 10.1016/s0014-5793(97)00605-4. [DOI] [PubMed] [Google Scholar]
- 140.Golub LM, Ramamurthy NS, McNamara TF, Greenwald RA, Rifkin BR. Tetracyclines inhibit connective tissue breakdown: new therapeutic implications for an old family of drugs. Crit Rev Oral Biol Med. 1991;2:297–321. doi: 10.1177/10454411910020030201. [DOI] [PubMed] [Google Scholar]
- 141.Yrjanheikki J, Tikka T, Keinanen R, Goldsteins G, Chan PH, Koistinaho J. A tetracycline derivative, minocycline, reduces inflammation and protects against focal cerebral ischemia with a wide therapeutic window. Proc Natl Acad Sci U S A. 1999;96:13496–13500. doi: 10.1073/pnas.96.23.13496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Pruzanski W, Greenwald RA, Street IP, Laliberte F, Stefanski E, Vadas P. Inhibition of enzymatic activity of phospholipases A2 by minocycline and doxycycline. Biochem Pharmacol. 1992;44:1165–1170. doi: 10.1016/0006-2952(92)90381-r. [DOI] [PubMed] [Google Scholar]
- 143.Zhang L, Kitaichi K, Fujimoto Y, Nakayama H, Shimizu E, Iyo M, Hashimoto K. Protective effects of minocycline on behavioral changes and neurotoxicity in mice after administration of methamphetamine. Prog Neuropsychopharmacol Biol Psychiatry. 2006;30:1381–1393. doi: 10.1016/j.pnpbp.2006.05.015. [DOI] [PubMed] [Google Scholar]
- 144.Tanibuchi Y, Shimagami M, Fukami G, Sekine Y, Iyo M, Hashimoto K. A case of methamphetamine use disorder treated with the antibiotic drug minocycline. Gen Hosp Psychiatry. 2010;32:559, e551–553. doi: 10.1016/j.genhosppsych.2009.12.005. [DOI] [PubMed] [Google Scholar]
- 145.Fujita Y, Kunitachi S, Iyo M, Hashimoto K. The antibiotic minocycline prevents methamphetamine-induced rewarding effects in mice. Pharmacol Biochem Behav. 2012;101:303–306. doi: 10.1016/j.pbb.2012.01.005. [DOI] [PubMed] [Google Scholar]
- 146.Snider SE, Hendrick ES, Beardsley PM. Glial cell modulators attenuate methamphetamine self-administration in the rat. Eur J Pharmacol. 2013;701:124–130. doi: 10.1016/j.ejphar.2013.01.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Mizoguchi H, Takuma K, Fukakusa A, Ito Y, Nakatani A, Ibi D, Kim HC, Yamada K. Improvement by minocycline of methamphetamine-induced impairment of recognition memory in mice. Psychopharmacology (Berl) 2008;196:233–241. doi: 10.1007/s00213-007-0955-0. [DOI] [PubMed] [Google Scholar]
- 148.Attarzadeh-Yazdi G, Arezoomandan R, Haghparast A. Minocycline, an antibiotic with inhibitory effect on microglial activation, attenuates the maintenance and reinstatement of methamphetamine-seeking behavior in rat. Prog Neuropsychopharmacol Biol Psychiatry. 2014;53:142–148. doi: 10.1016/j.pnpbp.2014.04.008. [DOI] [PubMed] [Google Scholar]
- 149.Boger HA, Middaugh LD, Granholm AC, McGinty JF. Minocycline restores striatal tyrosine hydroxylase in GDNF heterozygous mice but not in methamphetamine-treated mice. Neurobiol Dis. 2009;33:459–466. doi: 10.1016/j.nbd.2008.11.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Tata DA, Raudensky J, Yamamoto BK. Augmentation of methamphetamine-induced toxicity in the rat striatum by unpredictable stress: contribution of enhanced hyperthermia. Eur J Neurosci. 2007;26:739–748. doi: 10.1111/j.1460-9568.2007.05688.x. [DOI] [PubMed] [Google Scholar]
- 151.Northrop NA, Yamamoto BK. Persistent neuroinflammatory effects of serial exposure to stress and methamphetamine on the bloodbrain barrier. J Neuroimmune Pharmacol. 2012;7:951–968. doi: 10.1007/s11481-012-9391-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Asanuma M, Tsuji T, Miyazaki I, Miyoshi K, Ogawa N. Methamphetamine-induced neurotoxicity in mouse brain is attenuated by ketoprofen, a non-steroidal anti-inflammatory drug. Neurosci Lett. 2003;352:13–16. doi: 10.1016/j.neulet.2003.08.015. [DOI] [PubMed] [Google Scholar]
- 153.Nair A, Bonneau RH. Stress-induced elevation of glucocorticoids increases microglia proliferation through NMDA receptor activation. J Neuroimmunol. 2006;171:72–85. doi: 10.1016/j.jneuroim.2005.09.012. [DOI] [PubMed] [Google Scholar]
- 154.Perry VH. Stress primes microglia to the presence of systemic inflammation: implications for environmental influences on the brain. Brain Behav Immun. 2007;21:45–46. doi: 10.1016/j.bbi.2006.08.004. [DOI] [PubMed] [Google Scholar]
- 155.Northrop NA, Yamamoto BK. Cyclooxygenase activity contributes to the monoaminergic damage caused by serial exposure to stress and methamphetamine. Neuropharmacology. 2013;72:96–105. doi: 10.1016/j.neuropharm.2013.04.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Thomas DM, Kuhn DM. Cyclooxygenase-2 is an obligatory factor in methamphetamineinduced neurotoxicity. J Pharmacol Exp Ther. 2005;313:870–876. doi: 10.1124/jpet.104.080242. [DOI] [PubMed] [Google Scholar]
- 157.Thrash B, Thiruchelvan K, Ahuja M, Suppiramaniam V, Dhanasekaran M. Methamphetamine-induced neurotoxicity: the road to Parkinson’s disease. Pharmacol Rep. 2009;61:966–977. doi: 10.1016/s1734-1140(09)70158-6. [DOI] [PubMed] [Google Scholar]
- 158.Maurice T, Su TP. The pharmacology of sigma-1 receptors. Pharmacol Ther. 2009;124:195–206. doi: 10.1016/j.pharmthera.2009.07.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Yasui Y, Su TP. Potential molecular mechanisms on the role of the sigma-1 receptor in the action of cocaine and methamphetamine. J Drug Alcohol Res. 2016:5. doi: 10.4303/jdar/235970. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Nguyen EC, McCracken KA, Liu Y, Pouw B, Matsumoto RR. Involvement of sigma (sigma) receptors in the acute actions of methamphetamine: receptor binding and behavioral studies. Neuropharmacology. 2005;49:638–645. doi: 10.1016/j.neuropharm.2005.04.016. [DOI] [PubMed] [Google Scholar]
- 161.Itzhak Y. Repeated methamphetamine-treatment alters brain sigma receptors. Eur J Pharmacol. 1993;230:243–244. doi: 10.1016/0014-2999(93)90810-5. [DOI] [PubMed] [Google Scholar]
- 162.Ujike H, Kanzaki A, Okumura K, Akiyama K, Otsuki S. Sigma (sigma) antagonist BMY 14802 prevents methamphetamine-induced sensitization. Life Sci. 1992;50:PL129–134. doi: 10.1016/0024-3205(92)90466-3. [DOI] [PubMed] [Google Scholar]
- 163.Matsumoto RR, Shaikh J, Wilson LL, Vedam S, Coop A. Attenuation of methamphetamineinduced effects through the antagonism of sigma (sigma) receptors: Evidence from in vivo and in vitro studies. Eur Neuropsychopharmacol. 2008;18:871–881. doi: 10.1016/j.euroneuro.2008.07.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Takahashi S, Miwa T, Horikomi K. Involvement of sigma 1 receptors in methamphetamine-induced behavioral sensitization in rats. Neurosci Lett. 2000;289:21–24. doi: 10.1016/s0304-3940(00)01258-1. [DOI] [PubMed] [Google Scholar]
- 165.Stefanski R, Justinova Z, Hayashi T, Takebayashi M, Goldberg SR, Su TP. Sigma1 receptor upregulation after chronic methamphetamine self-administration in rats: a study with yoked controls. Psychopharmacology (Berl) 2004;175:68–75. doi: 10.1007/s00213-004-1779-9. [DOI] [PubMed] [Google Scholar]
- 166.Gekker G, Hu S, Sheng WS, Rock RB, Lokensgard JR, Peterson PK. Cocaine-induced HIV-1 expression in microglia involves sigma-1 receptors and transforming growth factor-beta1. Int Immunopharmacol. 2006;6:1029–1033. doi: 10.1016/j.intimp.2005.12.005. [DOI] [PubMed] [Google Scholar]
- 167.Shen K, Zhang Y, Lv X, Chen X, Zhou R, Nguyen LK, Wu X, Yao H. Molecular mechanisms involving sigma-1 receptor in cell apoptosis of bv-2 microglial cells induced by methamphetamine. CNS Neurol Disord Drug Targets. 2016;15:857–65. doi: 10.2174/1871527315666160518122816. [DOI] [PubMed] [Google Scholar]
- 168.Cuevas J, Rodriguez A, Behensky A, Katnik C. Afobazole modulates microglial function via activation of both sigma-1 and sigma-2 receptors. J Pharmacol Exp Ther. 2011;339:161–172. doi: 10.1124/jpet.111.182816. [DOI] [PubMed] [Google Scholar]
- 169.Matsumoto RR, Bowen WD, Su TP. Sigma receptors: chemistry, cell biology and clinical implications. Springer Science & Business Media. 2007 [Google Scholar]
- 170.Robson MJ, Turner RC, Naser ZJ, McCurdy CR, O’Callaghan JP, Huber JD, Matsumoto RR. SN79, a sigma receptor antagonist, attenuates methamphetamine-induced astrogliosis through a blockade of OSMR/gp130 signaling and STAT3 phosphorylation. Exp Neurol. 2014;254:180–189. doi: 10.1016/j.expneurol.2014.01.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Zhang Y, Lv X, Bai Y, Zhu X, Wu X, Chao J, Duan M, Buch S, Chen L, Yao H. Involvement of sigma-1 receptor in astrocyte activation induced by methamphetamine via up-regulation of its own expression. J Neuroinflammation. 2015;12:29. doi: 10.1186/s12974-015-0250-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.van Vliet AR, Verfaillie T, Agostinis P. New functions of mitochondria associated membranes in cellular signaling. Biochim Biophys Acta. 2014;1843:2253–2262. doi: 10.1016/j.bbamcr.2014.03.009. [DOI] [PubMed] [Google Scholar]
- 173.Huang Z, Liu S, Zhang L, Salem M, Greig GM, Chan CC, Natsumeda Y, Noguchi K. Preferential inhibition of human phosphodiesterase 4 by ibudilast. Life Sci. 2006;78:2663–2668. doi: 10.1016/j.lfs.2005.10.026. [DOI] [PubMed] [Google Scholar]
- 174.Suzumura A, Ito A, Yoshikawa M, Sawada M. Ibudilast suppresses TNFalpha production by glial cells functioning mainly as type III phosphodiesterase inhibitor in the CNS. Brain Res. 1999;837:203–212. doi: 10.1016/s0006-8993(99)01666-2. [DOI] [PubMed] [Google Scholar]
- 175.Mizuno T, Kurotani T, Komatsu Y, Kawanokuchi J, Kato H, Mitsuma N, Suzumura A. Neuroprotective role of phosphodiesterase inhibitor ibudilast on neuronal cell death induced by activated microglia. Neuropharmacology. 2004;46:404–411. doi: 10.1016/j.neuropharm.2003.09.009. [DOI] [PubMed] [Google Scholar]
- 176.Kiebala M, Maggirwar SB. Ibudilast, a pharmacologic phosphodiesterase inhibitor, prevents human immunodeficiency virus-1 Tatmediated activation of microglial cells. PLoS One. 2011;6:e18633. doi: 10.1371/journal.pone.0018633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Estes ML, McAllister AK. Alterations in immune cells and mediators in the brain: it’s not always neuroinflammation! Brain Pathol. 2014;24:623–630. doi: 10.1111/bpa.12198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Thomas DM, Dowgiert J, Geddes TJ, Francescutti-Verbeem D, Liu X, Kuhn DM. Microglial activation is a pharmacologically specific marker for the neurotoxic amphetamines. Neurosci Lett. 2004;367:349–354. doi: 10.1016/j.neulet.2004.06.065. [DOI] [PubMed] [Google Scholar]