Abstract
The pervasive and devastating nature of substance use disorders underlies the need for the continued development of novel pharmacotherapies. We now know that glia play a much greater role in neuronal processes than once believed. The various types of glial cells (e.g., astrocytes, microglial, oligodendrocytes) participate in various functions that are crucial to healthy central nervous system function. Drugs of abuse have been shown to interact with glia in ways that directly contribute to the pharmacodynamic effects responsible for their abuse potential. Through their effect upon glia, drugs of abuse also alter brain function resulting in behavioral changes associated with substance use disorders. Therefore, drug-induced changes in glia and neuroinflammatory processes have been investigated to treat various aspects of drug abuse and dependence. This manuscript presents a brief overview of the effects of each of the major classes of addictive drugs on glia. Next, the paper reviews the pre-clinical and clinical studies assessing the effects that glial modulators have on abuse-related behavioral effects, such as pleasure, withdrawal, and motivation. There is a strong body of pre-clinical literature demonstrating the general effectiveness of several glia-modulating drugs in models of reward and relapse. Clinical studies have also yielded promising results, though not as robust. There is still much to disentangle regarding the integration between addictive drugs and glial cells. Improved understanding of the relationship between glia and the pathophysiology of drug abuse should allow for more precise exploration in the development and testing of glial-directed treatments for substance use disorders.
1. The Need for Continued Discovery of Medications to Treat Substance Use Disorders
Drug abuse is a chronic, relapsing disease characterized by dysfunction in brain reward-, motivation-, and memory-related circuitry [1]. Compulsive drug seeking and use, despite harmful consequences, results in progressive disability and can lead to premature mortality [2]. Substance use disorders (SUDs) contribute to the deaths of millions of people each year directly through overdose or indirectly by worsening comorbid psychiatric and medical conditions. Drug abuse also aids the spread of infectious diseases, such as HIV and Hepatitis C, and is associated with crime and violence [3]. According to the most recent World Drug Report, globally, an estimated ~164 million people suffer from an alcohol or drug use disorder, meaning that their drug use is sufficiently harmful to necessitate treatment [3].
Worldwide, an estimated 27 million people suffered from opioid use disorder (OUD) in 2017 [3]. Medications to treat OUD have a well-established efficacy, improving quality of life and reducing premature mortality [4; 5]. Though long-term maintenance on μ-opioid receptor agonist (methadone), partial agonist (buprenorphine) or antagonist (naltrexone) medications have significant clinical utility, retention rates at six months are typically below 50% [6].
Like opioids, there are medication options available to the estimated 1.4% of the global population that suffer from alcohol use disorder [7]. First-line pharmacotherapies include the opioid receptor antagonist, naltrexone, and glutamatergic modulator, acamprosate [8]. Naltrexone has been shown to reduce heavy drinking but there is little evidence that it has significant effects on alcohol abstinence [9]. Meanwhile, acamprosate only increases the rate of alcohol abstinence by about 10% [10]. Second-line agents, such as disulfiram - and those not approved by the U.S. Food and Drug Administration (FDA), like topiramate and baclofen - have been shown to reduce alcohol consumption in clinical trials, but without significant effects on overall abstinence [8].
There are more treatment modalities for nicotine dependence (i.e., smoking cessation, tobacco use) than opioids and alcohol combined. There are two pharmacotherapies approved by the FDA for nictoine dependence - bupropion (norepinephrine and dopamine reuptake inhibitor) and varenicline (partial agonist of the nicotinic acetylcholine receptor) - and five forms of nicotine-replacement therapy (i.e., patch, gum, lozenge, inhaler and nasal spray)[11]. However, with all these treatments, the likelihood of long-term abstinence remains low [12; 13]. With an estimated 1.1 billion active tobacco users, the morbidity associated with tobacco use remains one of the largest global public health epidemics [14; 7]
There are no FDA-approved pharmacotherapies to assist the estimated 18.2 million cocaine users, or the 34.2 million non-medical users of amphetamine-type stimulants (e.g., methamphetamine, amphetamine, and 3,4-methylenedioxymethamphetamine (MDMA) or ‘Ecstasy’)[3]. These potent psychostimulants are among the most addictive drugs classes. Similarly, there are no pharmacotherapies available to treat cannabis use disorder, which may develop in as much as 10% of the estimated 192 million users [15,16, 3].
In sum, many efficacious and effective pharmacological interventions for alcohol and drug dependence exist. However, there remains significant room to improve rates of long-term abstinence. Additionally, there are several drugs with significant potential for abuse for which there are no pharmacotherapies with reliable and robust evidence of treatment efficacy in clinical trials. Therefore, there is a clear need for additional therapeutic strategies to help those who suffer from SUDs.
2. Glia: Much More Important Than Once Believed
Glia were once considered to be passive neuronal support, simply ensuring that neurons could function properly [17]. However, we now know that glia are actively involved in the development and protection of, and are critical to, healthy CNS function [18–20]. Glia constitute over 90% of the cells in the CNS and between 33 to 66% of the total brain mass - depending upon the mammalian species being examined [21,22]. It is now widely-recognized that glia play a significant role in many neuronal processes, such as neurotransmitter modulation, along with synaptic development and plasticity [23,24]. There are a variety of subtypes of glial cells in the CNS, with microglia, and the various macroglia (i.e. astrocytes, ependymal cells, oligodendrocytes, and radial cells) each having a specialized function [25].
Microglia are the brain’s immune cells, functioning to cleanse the extracellular fluid in order to maintain central homeostasis [26]. As the resident macrophage cells, microglia act as the first and main form of active immune defense in the CNS [27] Microglial processes directly interact with neurons, other glia types and blood vessels [28]. Microglia have been observed to have receptors for neurotransmitters, allowing them to respond to neuronal activation [29,30].
Astrocytes are a type of macroglia that control the level of neurotransmitter around synapses by regulating the concentrations of important ions and providing metabolic support [31,32]. Astrocytes also connect the vasculature and neurons by regulating local blood flow in response to neuronal activity and providing energy substrates (e.g., glucose) to neurons [33,17]. Astrocytes also release numerous gliotransmitters [e.g., glutamate and adenosine triphosphate (ATP)] known to regulate many CNS functions including synaptic plasticity, neural network activity and sleep [34]
Oligodendrocytes produce and maintain the myelin sheath that surrounds axons, allowing for rapid saltatory propagation of action potentials [35, 17]. Oligodendrocytes also release several neurotrophins, including nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), and neurotrophin-3 [36]. Relative to the other types of glia, little is known of the function of ependymal cells. We do know that ependymal cells line the spinal cord and ventricles of the brain and are involved in creating cerebrospinal fluid [37]. Finally, radial glia are progenitor cells that can generate neurons, astrocytes, and oligodendrocytes [38, 39].
Current SUD medication strategies focus on neuronal mechanisms underlying addiction. Common medication targets include the neurobiological substrates responsible for the rewarding and reinforcing properties of drugs of abuse, such as dopamine [40–43]. All drugs of abuse are believed to exert their reinforcing effects by increasing the transmission of dopamine within the nucleus accumbens (NAcc), achieved through various molecular pathways for each drug class [44]. Recently, however, drug and neuroimmune interactions have emerged as potential targets for treating SUDs. There is an extensive body of literature to suggest that glial cells are influenced by drugs of abuse and these interactions contribute to their acute and chronic behavioral effects [45, 46]. Therefore, the involvement of glia in the development and maintenance of SUDs has important implications for the development of novel pharmacotherapies. The majority of this research has focused on interactions between addictive drugs and microglia, astrocytes or their gliotransmitters [47].
The purpose of the current narrative review is to summarize the pre-clinical and clinical findings from studies that have sought to evaluate the potential of glial-modulating drugs to treat features of SUDs. Several experimental paradigms have been developed to screen medications for clinical utility in treating SUD. A primary focus of these paradigms is the ability of the medication to alter the abuse potential of drugs as quantified by attenuation of the drug’s rewarding and/or reinforcing effects [48]. “Reward” is conceptualized as the experienced enjoyment or arousal in response to the drug. “Reinforcement” is drug-seeking and drug-taking behaviors thought to reflect the physiological, behavioral, and cognitive drug effects. Other important experimental measures focus on other aspects of the addictive process, such as craving (subjective desire for the drug) and relapse (reinstatement of drug-seeking or drug-taking following a period of abstinence).
The review is organized by drug class (i.e., alcohol, cannabis, opioids, nicotine, and psychostimulants). First, the manuscript will briefly detail how each drug class interacts with glia and how these effects contribute to their behavioral pathology. Next, we review pre-clinical studies that have evaluated the effects of medications with glial-modulating properties on the pharmacodynamic effects of drugs of abuse (e.g., rewarding and reinforcing effects). Finally, the author reviews clinical trials that have evaluated the effects of glia modulators on addiction-related outcomes.
Using PubMed and PsychINFO, the author searched for English-language articles published between 1970 and 2019. The search included various combinations of the following terms: glia, astrocytes, microglia, addiction, substance use disorder, cocaine, alcohol, amphetamine, methamphetamine, heroin, opioids, nicotine, and marijuana. Over 500 publications were identified. After removing duplicates, titles and/or abstracts were reviewed to ensure relevance. The search criteria included original reports assessing the interaction between glia, glial products and drugs of abuse, or assessment of medications that act on glia as potential treatments for SUDs. Data from approximately 200 articles are included in the current review.
3. Drugs of Abuse and Neuroimmune Interactions as Targets for Medication Development
3.1. Opioid Effects on Glia
Glia have been shown to express all three opioid receptors subtypes (μ, ĸ, δ) and the mRNA that encodes them [49–51]. Glia μ-opioid receptor expression can account for 2–9% of the total observed in the CNS, with δ-opioid receptor glial expression estimated at 5% [52–55]. Though we know that glial ĸ-opioid receptor expression has been observed, its degree has yet to be quantified [56–57]. The endogenous μ, ĸ, and δ opioid receptors ligands (β-endorphin, enkephalins, and dynorphins, respectively) are synthesized in glia. However, little is known regarding the storage and release of endogenous opioid neuropeptides from glial cells [58].
Concerning exogenous opioids, both natural opiates (e.g., morphine and codeine) as well as synthetic and semi-synthetic opioids (e.g., oxycodone and heroin) have been shown to have effects on glia activity [59–60]. Pharmacologically and pharmacodynamically, morphine is commonly considered to be the archetypal μ-opioid receptor agonist (Mello and Bayer, 1998; Pathan and Williams 2012). As such, morphine is commonly used in pre-clinical investigations of opioids effects. Morphine administration increases the expression of cellular markers associated with glial cell activation, such as microglial CD11b (the β-integrin marker of microglia) and glial fibrillary acidic protein (GFAP), a hallmark intermediate filament protein in astrocytes [63–67].
The effect of morphine on GFAP is not observed when it is co-administered with the non-selective opioid receptor antagonist, naltrexone [63]. Morphine administration can also lead to increased expression of immune-modulating factors released from glial, such as cytokines: interleukin-1 beta (IL-1 β) and tumor necrosis factor-alpha (TNF-α), and chemokines: C-C motif chemokine ligand-5 (CCL5), and monocyte chemoattractant protein-1 (MCP-1) [68–72]. Morphine also enhances microglial migration and apoptosis [73–75].
Opioids are believed to interact with glia through the pattern recognition receptor, toll-like receptor-4 (TLR-4), a key mediator of innate immunity and inflammation [76–77]. TLR-4 is widely expressed in all glia types [78, 79]. Morphine binds to the myeloid differentiation factor 2 (MD-2), which induces TLR-4 oligomerization and results in glial activation [66, 80]. TLR-4 has been found to be required for many opioid-induced effects on glia [75,76]. Genetic knockout of TLR-4/MD-2 suppressed morphine-induced glial activation [81].
Concerning addictive processes, opioid actions upon glial cells also contribute to opioid actions within the brain’s reward center [82]. The mesolimbic dopamine pathway transmits dopamine from the ventral tegmental area (VTA) to the NAcc, and to components of the limbic system associated with emotion and memory formation [44, 83, 84]. Narita and Colleagues (2006) published one of the most robust studies to observe the direct actions of glia on the rewarding effects of drugs of abuse. The investigators found that intra-NAcc and intra-cingulate cortex administration of astrocyte-related soluble factors enhanced morphine conditioned place preference (CPP). Conditioned Place Preference is a pre-clinical, Pavlovian conditioning model commonly used to quantify the rewarding effects of drugs by spatially and temporally pairing distinct environments with distinct drug or non-drug states. The development of a preference for the drug-paired environment is considered to be an index of the rewarding value of the drug [85]. Conversely, microglia-related soluble factors failed to alter the rewarding effects of morphine. However, other studies suggest that opioid interactions with microglia contribute to opioid actions within the brain’s reward system. Inhibition of microglial activity has been shown to attenuate morphine-induced dopamine release in the NAcc and hinder the development of morphine CPP [86, 87]. Genetic manipulation of the TLR-4 receptor has provided further evidence of the relationship between opioid effects on glia and its behavioral outcomes. For example, TLR-4 knockout mice showed impaired development of CPP to morphine and oxycodone [66,88].
3.2. Glial Modulators as Potential Treatments of Opioid Use Disorder
As reviewed above, opioid-induced activation of glia appears to contribute significantly to the behavioral effects attributed to their misuse. As such, there has been interest in evaluating the potential of glial modulators as treatments for OUD.
Pre-clinical Studies:
Building upon research demonstrating the importance of TLR-4 modulation of the rewarding effects of opioids, researchers found that the low-affinity pharmacological TLR-4 inhibitor, (+)-naloxone, impairs the development of morphine CPP and the self-administration of remifentanil - a potent synthetic opioid. (+)-Naloxone also significantly attenuates morphine-induced dopamine release in the NAcc [88]. Furthermore, chronic antagonism of TLR-4 with (+)-naltrexone blocks cue-induced reinstatement of heroin self-administration [89]. Though compelling, recent work has failed to replicate the finding with (+)-naloxone and (+)-naltrexone [90]. This lack of replication could question the hypothesized role of TLR-4, procedural differences between the two investigations may be to responsible.
Other glial-modulating drugs have shown more consistent results across preclinical studies. Bland and Colleagues [86] investigated the ability of ibudilast, a non-selective phosphodiesterase (PDE) and TNF-α inhibitor [91], to reduce morphine-induced dopamine release. The co-administration of ibudilast (AKA, AV-411) with morphine significantly attenuated morphine-induced dopamine transmission within the reward pathway. In another study, ibudilast also reduced the somatic signs of opioid withdrawal [92], a contributor to the maintenance of OUD [93].
The ability of glia-modulating drugs to reduce the abuse potential of opioids, has also been observed in behavioral models. Propentofylline is a PDE inhibitor that hinders induced TNF-α released and increased the expression of the astroglial glutamate transporter (GLT-1) [94–96]. Propentofylline has been shown to suppress the development of morphine CPP [97]. Fluorocitrate is a metabolic inhibitor of glia that has also been shown to reduce morphine CPP [98,99]. In this study, fluorocitrate also reduced the signs of opioid withdrawal. Another glia modulator that has been investigated in this capacity is the microglia inhibitor, minocycline [100]. Direct administration of minocycline into the rat NAcc blocked the maintenance of morphine CPP and the reinstatement of morphine-seeking behavior – a pre-clinical model of relapse [101]. Finally, pioglitazone, which inhibits the expression of cytokines by microglia [102], attenuates opioid withdrawal, heroin self-administration, and heroin-induced dopamine release [103–106].
There is a robust body of pre-clinical literature to suggest that opioid effects on glia contribute to other important pharmacodynamic effects of opioids [107]. Cumulating evidence has demonstrated that the pro-inflammatory effects of exogenous opioids contribute to opioid-induced hyperalgesia and the development of opioid tolerance and dependence [108–109]. Ibudilast, minocycline, propentofylline, and fluorocitrate have all shown promise to increase the clinical utility of opioid drugs by improving their analgesic efficacy or antagonizing the development of morphine tolerance and/or hyperalgesia [65,92,110–115]. Procyanidins are a structurally diverse set of compounds that can suppress morphine-induced activation of microglia and other glial markers [116]. Procyanidins have also been found to attenuate morphine-induced antinociceptive tolerance [117]. Finally, statins are potent inhibitors of various glial pathways and products [118–120]. Simvastatin, rosuvastatin, and atorvastatin have all been shown to have significant protective effects against the development of tolerance to the antinociceptive effects of morphine, and physiological dependence [121–126]
In addition to direct actions, the downstream effects of opioid-induced changes in glial function may provide another means to pharmacologically alter the behavioral effects of opioids. Opioid effects on astrocytic glutamate transporters can lead to deficits in glutamate transmission [127]. Pharmacologically restoring these deficits in glutamate function has been shown to attenuate morphine CPP [128], reinstatement of hydromorphone CPP [129], as well as heroin-seeking behavior [130, 131]. N-acetylcysteine (NAC) is a drug that increases the expression and function of the cysteine-glutamate exchanger (xCT) and the glutamate transporter-1 (GLT-1) [132]. In rats, NAC treatment has been reported to disrupt cue-induced heroin seeking [133]. Table 1 summarizes the pre-clinical and clinical findings of the effects of glial modulators on the abuse potential of opioids.
Table 1.
Effects of Glial Modulators on the Abuse Potential of Opioids
| Preclinical Studies | |||||
|---|---|---|---|---|---|
| Glial Modulator | Mechanism of Action | Dose | Behavioral Outcome | Result | Citation |
| Propentofylline | Phosphodiesterase (PDE) inhibitor & Glutamate Transporter-1 (GLT-1) Upregulator | 3 μM | Rewarding Effects | ↓ | Narita et al., 2006 |
| Pioglitazone | Peroxisome Proliferator-Activated Receptor-Gamma (PPARγ) Activator & Cytokine Inhibitor | 60 mg/kg | Reinforcing Effects | ↓ | de Guglielmo et al., 2015 |
| 30 mg/kg | Reinstatement of Drug-Seeking | ↓ | de Guglielmo et al., 2017 | ||
| 30 mg/kg | Withdrawal or Physiological Dependence | ↓ | |||
| Ceftriaxone | Cephalosporin Antibiotic & Upregulator of GLT-1 and Cystine/Glutamate Antiporter (xCT) | 200 mg/kg | Reinstatement of Drug-Seeking | ↓ | Shen et al., 2014 |
| 200 mg/kg | Rewarding Effects | ↓ | Alshehri et al., 2018 | ||
| Fluorocitrate | Glial Metabolic Inhibitor | 1 nmol/1μl | Rewarding Effects | ↓ | Seyedaghamiri et al., 2018 |
| Minocycline | Tetracycline Antibiotic & Microglial Inhibitor | 50 mg/kg | Rewarding Effects | ↓ | Hutchinson et al., 2008 |
| 50 mg/kg | Withdrawal or Physiological Dependence | ↓ | Hutchinson et al., 2009 | ||
| 10 & 20 μg | Rewarding Effects | ↓ | Zhang et al., 2012 | ||
| 1, 5, & 10 μg/0.5 μL | Reinstatement of Drug-Seeking | ↓ | Arezoomandan and Haghparast, 2016 | ||
| (+)-Naloxone | Non-Opioid Isomer of the Opioid Receptor Antagonist & Toll-Like Receptor-4 (TLR-4) Antagonist | 1 mg/kg | Rewarding Effects | ↓ | Hutchinson et al., 2012 |
| 1 mg/kg | Reinforcing Effects | ↓ | |||
| (+)-Naltrexone | Non-Opioid Isomer of the Opioid Receptor Antagonist & TLR-4 Antagonist | 15 & 30 mg/kg | Reinforcing Effects | -- | Theberge et al., 2013 |
| 15 & 30 mg/kg | Reinstatement of Drug-Seeking | ↓ | |||
| (+)-Naloxone (+)-Naltrexone | Non-Opioid Isomer of the Opioid Receptor Antagonist & TLR-4 antagonist | 32 & 56 mg/kg | Reinforcing Effects | -- | Tanda et al., 2016 |
| Ibudilast | Phosphodiesterase and Tumor Necrosis Factor-Alpha (TNF-α) Inhibitor | 15 mg/kg | Withdrawal or Physiological Dependence | ↓ | Hutchinson et al., 2009 |
| N-Acetylcysteine | Upregulator of GLT-1 & Glial Cystine-glutamate Exchange Activator | 90 mg/kg | Reinforcing Effects | ↓ | Hodebourg et al., 2019 |
| 100 mg/kg | Reinstatement of Drug-Seeking | ↓ | Zhou and Kalivas 2008 | ||
| MS-153 | Glial Glutamate Transport Activator | 10 mg/kg | Rewarding Effects | ↓ | Nakagawa et al., 2005 |
| Clinical Studies | |||||
| Glial Modulator | Mechanism of Action | Dose | Behavioral Outcome | Result | Citation |
| Minocycline | Tetracycline Antibiotic & Microglial Inhibitor | 100 & 200 mg | Positive Subjective Effects | ↓ | Mogali et al.,2014 |
| 200 mg | Craving | -- | Arout et al., 2018 | ||
| Ibudilast | PDE and TNF-α Inhibitor | 20 & 40 mg B.I.D | Withdrawal | ↓ | Cooper et al., 2016 |
| 20 & 40 mg B.I.D | Positive Subjective Effects | -- | Cooper et al., 2017 | ||
| 50 mg B.I.D | Positive Subjective Effects | ↓ | Metz et al., 2017 | ||
| 50 mg B.I.D | Reinforcing Effects | ↓ | |||
| 50 mg B.I.D | Craving, Reinstatement, or Relapse | ↓ | |||
| Pioglitazone | PPARγ Activator & Cytokine Inhibitor | 15 & 45mg | Positive Subjective Effects | – | Jones et al., 2016 |
| 45 mg | Craving | ↓ | Jones et al., 2018 | ||
| 45 mg | Positive Subjective Effects & Reinforcing Effects | -- | |||
Human Studies:
Clinical research is only beginning to replicate the promising pre-clinical findings concerning the treatment potential of glia-modulating drugs (see [134] and [135] for previous reviews) Cooper and Colleagues (2016) assessed the ability of ibudilast to alter opioid withdrawal symptoms among participants with OUD. Opioid-dependent volunteers (N=31) were maintained on morphine and then abruptly transitioned to placebo (0 mg). Participants were randomized to active (20 or 40 mg twice daily [BID]) or placebo ibudilast, while being maintained on morphine (120 mg/day) for 14 days, and subsequently placebo for the last seven days of the study. Opioid withdrawal was assessed daily using the Subjective and Clinical Opioid Withdrawal Scales (SOWS and COWS, respectively). All groups exhibited withdrawal during the third week. Analyses combining the two active ibudilast conditions found that this group had lower ratings of: “anxious,” “perspiring,” “restless,” and “stomach cramps” during the week in which they were withdrawing from morphine [136].
In a later publication, Cooper and Colleagues reported no “consistent” effects of ibudilast on the subjective effects of oxycodone [137]. Metz et al., (2017) extended these findings by testing the effects of ibudilast on the positive subjective and reinforcing effects of oxycodone. Opioid-dependent participants with OUD (N=11) first underwent in-patient detoxification followed by maintenance on ibudilast (0 and 50 mg BID, double-blind, randomized). The subjective and reinforcing effects of oral oxycodone (0, 15, and 30 mg/70 kg) were assessed under each ibudilast maintenance condition. While under active ibudilast maintenance, subjects’ ratings of drug “liking” were significantly decreased for the median oxycodone dose (15 mg). Oxycodone self-administration (15 mg) was also significantly reduced in the active ibudilast condition. Similar effects of ibudilast maintenance were found on the 30 mg dose of oxycodone, though they only approached statistical significance. Self-reported “craving” for opioids was also significantly reduced by ibudilast [138].
In a pilot study, Mogali et al. (2014) also tested the ability of a glial modulator to alter the positive subjective effects of oral oxycodone, along with opioid craving. Non-dependent recreational opioid users (N=7) participated in this randomized, double-blind outpatient study. Subjects completed five sessions in which they received acute minocycline (0, 100, or 200 mg) in combination with oral oxycodone (0, 40 mg). Compared to placebo, minocycline reduced participants’ ratings of “high,” drug “liking,” “good effect,” as well as how much participants reported that they “would pay” for the oxycodone dose they received [139]. In a later study among an opioid-dependent sample, minocycline (200 mg/day for 15 days) failed to alter outcomes, such as opioid craving and withdrawal [140]. However, the minocycline maintenance period did not significantly alter serum cytokine levels, therefore, the negative findings may be the result of an insufficient effect on glial activity.
In another investigation of dependent non-medical opioid users, Jones et al. (2018) assessed the ability of pioglitazone to alter the abuse potential of heroin. Heroin-dependent participants were recruited for a 3-week study. Upon admission, participants were randomized to either active (45 mg, n=14) or placebo (0 mg, N=16) pioglitazone maintenance for the duration of the study. After 5–7 days of stabilization on sublingual buprenorphine (8 mg, BID), participants began testing. Pioglitazone maintenance failed to alter the reinforcing or positive subjective effects of heroin, but did reduce heroin craving and overall anxiety [141].
The interaction between pioglitazone and oxycodone was also tested within a sample of non-dependent recreational prescription opioid users (N=17 completers). In this study participants were maintained for ~2 weeks on each of three doses of pioglitazone (0mg, 15mg, 45mg). Under each pioglitazone maintenance condition, participants completed a laboratory session assessing the subjective, analgesic, physiological and cognitive effects of oxycodone (0, 10, and 30 mg). Oxycodone produced typical μ agonist effects, such as miosis, decreased pain perception, and positive subjective effects. Yet, oxycodone effects did not vary as a function of the pioglitazone maintenance [142].
3.3. Psychostimulant Effects on Glia
Cocaine and amphetamine-type stimulants (e.g., d-amphetamine, methamphetamine, and MDMA) are potent psychostimulants whose abuse potential primarily results from their potent agonist effects upon monoamine transmission - particularly dopamine [143,144]. Among the cocaine-mediated secretory cytokines, upregulation of inflammatory factors, such as IL-1B, IL-6, and TNF-α has been demonstrated in both in vitro and in vivo studies [145–148]. There are numerous proposed mechanisms for cocaine-induced astrocytosis and microglial, though our understanding of them is incomplete [149–156].
Methamphetamine is also thought to have similar actions upon glia. Both cocaine and methamphetamine stimulate intracellular sigma-1 receptors, a means by which both drugs activate astrocytes and microglia [157–164]. Methamphetamine is thought to induce microglial apoptosis through this pathway [165]. Methamphetamine also differentially modulates the production of IL-6 and TNF-α in microglia [166,167].
Like opioids, TLR-4-induced microglial reactivity has been found to be critical to psychostimulant-induced dopamine release, reward, and reinforcement [156]. Of particular relevance may be the fact that many of the neuroinflammatory effects of cocaine, amphetamine, and methamphetamine are found within the brain’s reward center. The self-administration of several drugs of abuse has been found to alter gliogenesis in the medial prefrontal cortex [168], an area of the brain involved in reward processing, attention, and drug reinstatement [169]. In the NAcc, cocaine and methamphetamine increase the expression of glial fibrillary acidic protein (GFAP), an indicator of astrocyte activation GFAP [97,170–174]. Methamphetamine also increases the expression of the 27-kD heat shock protein (HSP27: a robust product of astrocytes and microglia) in the cortex, striatum, and hippocampus - all critical components of addiction neurobiology [175–177].
Region-specific, psychostimulant-induced activation of glia, has been associated with behavioral sensitization, whereby the behavioral response to a specified dose of drug increases with repeated administration [178]. Behavioral sensitization is commonly assessed in pre-clinical studies to model the impact of repeated drug exposure on behavior and neuroplasticity [179]. Studies have shown significant correlations between the degree of amphetamine-induced behavioral sensitization and the magnitude of their effects on astrocytes [170, 180, 181]. More specifically, increased GFAP immunoreactivity in the striatum and increased astrocyte proliferation/migration [indicated by increased expression of basic fibroblast growth factor (bFGF)] in the NAcc, have been shown to be direct contributors to the persistent behavioral sensitization seen with repeated exposure to psychostimulants [182–183].
Methamphetamine-induced increase in microglial activation in the reward center has been identified by increased immunoreactivity of the activated microglia marker, macrophage-1 antigen-CD11b (Mac1-CD11b). Methamphetamine effects on Mac1-CD11b have also been linked to drug-induced behavioral sensitization [175,184,185].
Dopamine depletion occurs following repeated exposure to psychostimulant drugs [186,187]. A direct association has been observed between the neurotoxic effect of methamphetamine on dopamine and its actions upon on glia [184]. Imaging studies in humans have confirmed that methamphetamine’s effects on glia are conserved across rodents and humans [188]. However, differences in the expression of activated microglia seen between human methamphetamine users relative to matched-controls, were not replicated with cocaine users [189].
In both pre-clinical and clinical studies, psychostimulants are neurotoxic to striatal dopamine [190]. This neurotoxicity is at least partly mediated through glia. Furthermore, neuroadaptations in midbrain VTA neurons, induced by psychostimulant effects on glial-derived neurotropic factor (GDNF), play an important role in craving and drug-seeking behavior [191–194]. In a human imaging study, glial activation also enhanced the striatal dopamine elevation resulting from the administration of the amphetamine-type stimulant, methylphenidate [195]. There is also a growing body of literature linking psychostimulant-induced disrupted communication between glia and neurons with the pathological learning involved in SUDs [196, 197]. Therefore, psychostimulant effects on glia may contribute directly to all of the key factors to psychostimulant addiction: drug liking, escalating use, craving, and relapse [198].
Psychostimulants also produce profound alterations in the ability of glia to regulate glutamate homeostasis. Psychostimulants produce increases in extracellular glutamate in the CNS [171; 181, 199- 201]. This effect can occur via psychostimulant actions on both microglia and astrocytes [147, 202, 203]. However, much of the research has focused on psychostimulant effects on the glutamate transporter (GLT-1) that is expressed on astrocytes, critical for clearing synaptic glutamate [204]. There is evidence that both psychostimulant exposure and extinction/abstinence contribute to alterations in astrocytic glutamate clearance [202, 205–207]. Glutamate is a neurotransmitter highly involved in the learning behaviors associated with SUDs [208]. Disruptions in glutamate homeostasis have been linked to the neural mechanisms underlying relapse. Glutamatergic hyper-excitability has also been observed in the cocaine- and stimulant-addicted brain [209].
3.4. Glial Modulators as Potential Treatments of Psychostimulant Use Disorder
Pre-clinical Studies:
Medications that can restore the ability of astrocytes to regulate glutamate homeostasis have been found to alter psychostimulant-induced behavioral and neuroadaptive effects. The glutamate transport activator, MS-153, attenuates cocaine- and methamphetamine-induced CPP [128]. Ceftriaxone is an antibiotic that also modulates the uptake activity of glial GLT-1. Chronic ceftriaxone treatment blocks cocaine- and methamphetamine-seeking behavior in rodents [210–215]. NAC is a drug that increases the expression and function of the cysteine-glutamate exchanger and the GLT-1 and restore psychostimulant-induced glutamate dysregulation [216–218]. In rodent models, NAC reduces cocaine-maintained responding, escalation of cocaine self-administration, and cocaine-seeking [131, 212, 219-224, though see [225]).
Surprisingly, N-acetylcysteine has not been shown to be as efficacious in rodent studies of methamphetamine, as it has been with cocaine. [226]. However, other glial modulators that have shown promise include the microglia inhibitor, minocycline. In mice, pretreatment with minocycline significantly reduced methamphetamine-induced dopamine release across species [185, 227]. Additionally, minocycline has been shown to have protective effects against methamphetamine-induced behavioral sensitization and deficits in long-term memory associated with repeated methamphetamine exposure, [185,228]. Minocycline has also been shown to attenuate the reinforcing effects of methamphetamine, and reinstatement of methamphetamine-seeking behavior [229, 230].
Minocycline also attenuates cocaine-induced dopamine release [155] and prevented the development of cocaine-induced sensitization [231]. However, with both cocaine and methamphetamine, minocycline could not reverse drug-induced sensitization, once established. The anti-inflammatory glial attenuator and non-selective PDE inhibitor, ibudilast, is able to attenuate cocaine sensitization [232]. Ibudilast also suppresses stress- and drug-induced reinstatement of methamphetamine seeking [233], locomotor sensitization [234] and self-administration [229]. Similarly, propentofylline (a PDE inhibitor/ GLT-1 upregulator) decreases preference for methamphetamine-associated cues [97] and inhibited cue-primed cocaine seeking [235]. There is also evidence that pharmacological inhibition of TLR-4 and PPARγ agonism (using GW9662) may alter psychostimulant effects responsible for initial use and relapse [155, 236, 237].
Human Studies:
The PDE and TNF-α inhibitor, pentoxifylline, was one of the first glial-modulator drugs tested for cocaine use disorder in a pilot clinical trial by Ciraulo and Colleagues [238]. Sixteen cocaine-dependent participants were maintained for 8 weeks on pentoxifylline (1200 mg/day). The investigators found a trend towards decreased cocaine use; the only positive medication effect observed among five medications tested.
Another group of investigators examined the effects of celecoxib maintenance (200 mg/day for 8 weeks) on cocaine use outcomes [239]. Celecoxib is a cyclooxygenase 2 (COX-2) inhibitor that counteracts glia-driven neuroinflammation [240]. No significant effect of celecoxib was found on cocaine abstinence rates, craving, or related psychosocial measures. Among 30 treatment-seeking adults with cocaine use disorder, pioglitazone (45 mg/ day) resulted in a significantly greater decrease in craving (vs placebo [241]). Pioglitazone treatment also improved brain white matter integrity that may be associated with impulsivity, decision making, and time to relapse [242, 243].
NAC is one of the most studied glial modulators for treating cocaine use disorder. First, in an open-label pilot study, 23 treatment-seeking cocaine-dependent patients received NAC at doses of 1200 mg/day, 2400 mg/day or 3600 mg/day for 4 weeks [244]. Higher doses of NAC (2400 and 3600 mg) increased retention in treatment and aided in reducing cocaine use. Concurrently, La Rowe and Colleagues (2007) reported that 3-day NAC treatment (600 mg) decreased cocaine desire during a cocaine cue-reactivity procedure (N=15) [245]. In another pilot study among cocaine-dependent males (N=4), NAC treatment (1200–2400 mg/day, 4 days) reduced cocaine craving following an intravenous cocaine injection [246].
Following up on their clinical laboratory data, La Rowe and Colleagues (2013) conducted a clinical trial among treatment-seeking adults with cocaine use disorder (N=111). All participants received cognitive-behavioral therapy over an 8-week treatment period and were randomized to receive either 600 or 1200 mg (twice per day) NAC or placebo during the treatment phase. Eight-week treatment with NAC failed to reduce cocaine use or craving. However, a positive signal was found for the effects of 1200 mg, showing lessened time to relapse and lower rating of drug craving [247].
Back and Colleagues (2016) tested NAC for post-traumatic stress disorder symptomology and substance abuse, among dual-diagnosed veterans (N=35). The investigators found that active NAC maintenance (2400 mg/day, for 8 weeks) significantly reduced drug craving and depression but had no effect on drug-taking behavior [248]. Most recently, Levi-Bolin et al., (2017) found that NAC maintenance (2400 mg) reduced the incentive salience of cocaine-related stimuli and has promising effects on cocaine self-administration [249]. Though clinical trials with NAC have an inconsistent pattern of results, a review and meta-analysis of the efficacy of NAC in treating SUDs found that in comparison to placebo, NAC was significantly superior in treating drug craving [250]. See also a review by Nocito-Echevarria and Colleagues (2017) [251].
In comparison to cocaine, there are fewer clinical studies examining glial modulators as a potential pharmacotherapy for the abuse of amphetamine-type stimulants. However, like cocaine, NAC has shown promise in reducing craving for methamphetamine. Significant reductions in craving were observed among methamphetamine–dependent volunteers (N=32) following 4 weeks of maintenance on NAC (1200 mg/day) [252].
In other clinical trials, ibudilast attenuated subjective effects of methamphetamine among methamphetamine-dependent volunteers (N=11) [253]. Participants received oral placebo, or ibudilast (40 mg, 100 mg BID) for 7 days. Following intravenous methamphetamine (15 and 30 mg), active ibudilast reduced subjective effects including: “high,” “good,” and “like.” In another clinical study (N=11), ibudilast (50 mg, BID) improved attention during early abstinence from methamphetamine among methamphetamine-dependent participants [254].
The promising pre-clinical findings with minocycline have also been replicated in clinical samples. Among volunteers (N=10) with no history of drug abuse, minocycline treatment (200 mg/day for 5 days) was found to attenuate the positive subjective effects of oral dextroamphetamine (20 mg/70 kg). Minocycline treatment did not have a significant effect on dextroamphetamine self-administration [255].
3.5. Alcohol Effects on Glia
The pharmacological effects of alcohol (or ethanol) are diverse and include alterations in Gamma-Aminobutyric Acid (GABA) and glutamate receptor function, endogenous opioids, and VTA dopamine activity [256]. Alcohol is believed to activate transcription factors that increase the expression of pro-inflammatory molecules [257–259]. Alcohol effects in the CNS are also associated with elevations in various innate immune signaling molecules, including IL-1β, IL-6, CCL3, CXCL2, GFAP, TLR-3,−4, and TNF-α [260–263]. Microglia and pro-inflammatory cytokines also alter synaptic signaling in a way that modulates alcohol effects on GABA receptors [45,264]
The effects of alcohol on glia have been shown to be dependent upon the dose and pattern of alcohol administration. For example, rats provided free access to alcohol for shorter periods (4–12 weeks) show increased activation of astrocytes (measured by GFAP), but decreased astrocyte activation was observed under conditions of extended alcohol access (36 weeks) [265, 266]. Glial response to alcohol also appears to be dependent upon the brain region assessed. Studies in rodent models of alcohol consumption have shown significant regional variability in the expression of pro-inflammatory molecules in the brain [266–271].
In humans with alcohol dependence, analysis of post-mortem tissue reveals reduced GFAP immunoreactivity in the dorsolateral prefrontal cortex, and a dramatic loss of astrocytes in the hippocampus [272,273]. Meanwhile, increased immunoreactivity for microglial-specific proteins is observed in the cingulate cortex of alcohol-dependent participants [274]. There is also evidence of increased expression of neuroinflammatory genes and protein markers in the frontal cortex [275–276].
Like all drugs of abuse covered so far, TLR-4 has been found to be a key receptor involved in alcohol-induced neuroimmune signaling [277]. Astrocytes lacking TLR-4, or its critical adaptor molecules, fail to show activation by alcohol [278]. Similar effects can be produced in vivo. For example, alcohol-induced expression of pro-inflammatory molecules was blocked by TLR-4 neutralizing antibodies [257]. Additionally, TLR-4 knockout mice fail to demonstrate evidence of alcohol-induced glial cell activation [257, 260, 279–284]. Mice lacking TLR-4 are also protected against alcohol anxiety-like behavior associated with alcohol withdrawal, suggestive of involvement of alcohol-neuroimmune interactions in the greater pathophysiology of alcoholism [278]. Direct manipulation of TLR-4 can also influence drinking behavior. Alcohol-preferring rat strains have been found to have increased expression of TLR-4 [285]. Viral-mediated knockdown of TLR-4 expression reduces alcohol consumption in rats [285–286]. Genetic perturbation of chemokine networks can also reduce drinking behavior (Blednov et al., 2005).
One of the more intriguing and novel effects of alcohol is its effects upon high-mobility group box 1 (HMGB1), an endogenous TLR-4 agonist [288]. Alcohol administration to mice and rats can lead to persistent increases in the expression of HMGB1 (along with various TLR receptors) and triggers innate immune responses through binding to TLR-4 [268, 270, 289]. Increases in HMGB1 and TLR signalizing have been correlated with the level of alcohol consumption among moderate and heavy drinkers [290]. Repeated exposure to alcohol has been shown to sensitize microglia toward hyperactivity in an HMGB1-dependent manner [291]. Sensitized or “primed” microglia produce a greater inflammatory response to immune stimuli than non-primed microglia [292]. Alcohol-induced neuroimmune sensitization is thought to be a significant contributor to the neurodegeneration seen with chronic alcohol use [266,293]. This sensitization is also thought to contribute to loss of control, increased impulsivity and negative affect, hallmarks of the pathology of alcohol use disorder [294, 295]. In humans with alcohol use disorder, plasma levels of pro-inflammatory cytokines have been shown to correlate with addiction severity and alcohol craving [296,297]. There is also a growing body of literature implicating neuroimmune systems in alcohol withdrawal [298–300].
3.6. Glial Modulators as Potential Treatments of Alcohol Use Disorder
Pre-clinical Studies:
Given the proposed molecular, cellular and behavioral contribution of neuroinflammation to alcohol use disorder, immune therapies may provide benefits for prevention and recovery. As previously reported with opioids and psychostimulants, the microglial inhibitor, minocycline, has demonstrated promise in pre-clinical studies. Minocycline has been shown to reduce alcohol self-administration and reinstatement of alcohol CPP [301–304]. Minocycline has also been shown to protect the developing brain against alcohol-induced damage [166]. Other antibiotics with glial-modulating effects, such as rapamycin, indomethacin, tigecycline and glycyrrhizin, have also show promise in pre-clinical studies [305–314]. Ceftriaxone, the glial GLT-1 modulator, has shown particular effectiveness in relapse models of drinking behavior [315–318].
PDE-4 inhibitors, including ibudilast, mesopram, rolipram, and CDP-840 have also been shown to decrease alcohol intake in rodents [319–322]. Meanwhile, microglial-inhibiting effects of pioglitazone have been found to reduce the toxicity of alcohol in pre-clinical models of fetal alcohol syndrome [323, 324]. Pioglitazone has also been shown to have protective effects against alcohol-induced neuronal and cognitive damage [325], suppressing alcohol drinking and relapse to alcohol seeking [326–329].
NAC offers promising therapeutic value to inhibit alcohol-induced adverse effects in rodents [330]. In Zebrafish, withdrawal effects following repeated alcohol exposure are blocked by NAC [331]. NAC is also able to reduce extinction responding and reacquisition of alcohol self-administration in rodents [332].
Human Studies:
Table 3 details candidate medications that have been found to be beneficial for alcohol use disorders in both pre-clinical and human studies. Unfortunately, there is a lack of clinical exploration of the medications that have shown promise in animal models. However, in a recent trial, ibudilast has shown potential utility in treating individuals with alcohol use disorder. Ray and Colleagues (2017) examined the effects of ibudilast (50 mg, BID) on subjective response to alcohol, drug cue and stress among participants with current alcohol use disorder (N=24). After 7 days of medication maintenance, participants completed an intravenous alcohol administration session, an alcohol cue session, and a stress-exposure session. Active ibudilast maintenance was associated with greater resilience in response to stress- and alcohol-cue exposure, reduced drug craving, and attenuation of some subjective effects of alcohol [333]. Minocycline has also been tested in clinical samples. Petrakis et Colleagues (2019) conducted a clinical trial in which heavy drinkers (N=48) were randomized to receive placebo, 100 mg, or 200 mg of minocycline for 10 days. The trial found no effect of either active dose of minocycline on subjective response to alcohol, alcohol-induced craving, or serum cytokine levels. The investigators therefore concluded that short-term minocycline maintenance may not alter alcohol-related inflammation or subjective response in humans [334].
Table 3.
Effects of Glial Modulators on the Abuse Potential of Alcohol
| Preclinical Studies | |||||
|---|---|---|---|---|---|
| Glial Modulator | Mechanism of Action | Dose | Behavioral Outcome | Result | Citation |
| Indomethacin | Cyclooxygenase-1&2 Inhibitor | 1.9, 2.5 & 5 mg/kg | Reinforcing Effects | ↓ | George, 1989 |
| Rolipram | Selective Phosphodiesterase-4 (PDE-4) Inhibitor | 0.25 & 0.5 mg/kg | Reinforcing Effects | ↓ | Hu et al., 2011 |
| 0.1 & 0.2 mg/kg | Reinforcing Effects | ↓ | Wen et al., 2012 | ||
| 1 mg/kg | Reinforcing Effects | ↓ | Blednov et al., 2014 | ||
| CDP840 | Selective PDE-4 Inhibitor | 10 & 25 mg/kg | Reinforcing Effects | ↓ | |
| Mesopram | Selective PDE-4 Inhibitor | 5 mg/kg | Reinforcing Effects | ↓ | |
| Piclamilast | Selective PDE-4 Inhibitor | 1 mg/kg | Reinforcing Effects | ↓ | |
| Propentofylline | PDE Inhibitor & Glutamate Transporter-1 (GLT-1) Upregulator | 5 mg/kg | Reinforcing Effects | -- | |
| Various | PDE-1,3,5 and Non-specific Inhibitors | -- | Reinforcing Effects | -- | |
| Ibudilast | PDE & Tumor Necrosis Factor-Alpha (TNF-α) | 3,6 & 9 mg/kg | Reinforcing Effects | ↓ | Bell et al., 2015 |
| Minocycline | Tetracycline Antibiotic & Microglial Inhibitor | 50 mg/kg | Reinforcing Effects | ↓ | Agrawal et al., 2011 |
| 60 & 80 mg/kg | Reinforcing Effects | ↓ | Syapin et al., 2016 | ||
| Rapamycin | T-Lymphocyte & Astrocyte Inhibitor | 50 & 100 ng/side | Reinforcing Effects | ↓ | Cozzoli et al., 2016 |
| Tigecycline | Glycylcycline Antibiotic & Microglial Inhibitor | 40, 60, 80 & 100 mg/kg | Reinforcing Effects | ↓ | Bergeson et al., 2016 |
| 60 & 80 mg/kg | Reinforcing Effects | ↓ | Syapin et al., 2016 | ||
| 80 mg/kg | Withdrawal | ↓ | Martinez et al., 2016 | ||
| Ceftriaxone | Cephalosporin Antibiotic & Upregulator of GLT-1 and Cystine/Glutamate Antiporter (xCT) | 100 mg/kg | Reinforcing Effects | ↓ | Alhaddad et al., 2014 |
| 100 mg/kg | Reinforcing Effects | ↓ | Das et al., 2015 | ||
| 100 mg/kg | Reinforcing Effects | ↓ | Sari et al., 2014 | ||
| 200 mg/kg | Reinforcing Effects | ↓ | Sari et al., 2016 | ||
| 50 & 100 mg/kg | Reinforcing Effects & Reinstatement of Drug Seeking | ↓ | Qrunfleh et al., 2013 | ||
| 200 mg/kg | Reinstatement of Drug Seeking | ↓ | Rao and Sari, 2014 | ||
| 100 mg/kg | Reinstatement of Drug Seeking | ↓ | Weiland et al., 2015 | ||
| 200 mg/kg | Reinforcing Effects | ↓ | Lee et al., 2013 | ||
| (+)-Naloxone | Non-Opioid Isomer of the Opioid Receptor Antagonist & Toll-Like Receptor-4 (TLR-4) Antagonist | 30 & 60 mg/kg | Reinforcing Effects | -- | Harris et al., 2017 |
| Pioglitazone | Peroxisome Proliferator-Activated Receptor-Gamma (PPARγ) Activator & Cytokine Inhibitor | 30 mg/kg | Reinstatement of Drug Seeking | ↓ | Blednov et al., 2015 |
| 10 & 30 mg/kg | Reinstatement of Drug Seeking | ↓ | Stopponi et al., 2013 | ||
| Clinical Studies | |||||
| Glial Modulator | Mechanism of Action | Dose | Behavioral Outcome | Result | Citation |
| Ibudilast | PDE & TNF-α Inhibitor | 50 mg | Positive Subjective Effects | -- | Ray et al., 2017 |
| 50 mg | Craving | ↓ | |||
| Minocycline | Tetracycline Antibiotic & Microglial Inhibitor | 100 & 200 mg | Positive Subjective Effects | -- | Petrakis et al., 2019 |
3.7. Nicotine Effects on Glia
Due to the hundreds of chemical constituents of tobacco, it has been difficult to specify the effects of nicotine on glia [335]. We do know that smoking promotes the release of pro-inflammatory signals in a dose-dependent manner [336]. However, nicotine itself is thought to be immunosuppressive [337]. In culture, nicotine inhibits microglial proliferation through direct effects on nicotinic acetylcholine receptors (nAChRs) that are expressed on microglia [338–341]. nAChRs are widely distributed throughout the CNS and are expressed on several types of non-neuronal cells [338]. nAChR activation has been shown to have immunosuppressive properties [339] and in vitro models have also shown that long-term nicotine treatment suppresses the reactivity of astrocytes [342]. In humans, direct comparison of smokers to non-smokers using positron emission tomography (PET) found indicators of less microglia activation, the magnitude of which was inversely correlated with the number of cigarettes smoked per day [343]. There is also evidence that long-term exposure to nicotine and/or other chemicals found in tobacco smoke impairs the functions of oligodendrocytes [344–345].
Non-neuronal effects of nicotine are thought to enhance hippocampal synaptic transmission and long-term memory [346]. Nicotine effects on glia have also been shown to have neuroprotective effects on dopamine, and modulate molecular measures in the NAcc, substantia nigra and frontoparietal cerebral cortex [347–350]. Therefore, there is a substantial body of evidence to implicate the interaction between nicotine and glia in the development and progression of nicotine abuse.
Pre-clinical Studies:
Concerning the utility of using glia modulators for the treatment of nicotine use, there have been relatively few studies. The glial GLT-1 modulator, ceftriaxone, has been the most extensively studied in pre-clinical models. Ceftriaxone has been shown to reduce acquisition and reinstatement of nicotine CPP, nicotine intake and withdrawal [351–353]. NAC has shown similar effects. In mice, NAC decreased nicotine’s rewarding effects [354]. In rats, NAC also reduced nicotine self-administration and cue-induced reinstatement of nicotine seeking [355, 356], reduced nicotine withdrawal [357], and enhanced extinction of nicotine-associated cues [358]. Like some studies above, Powell and Colleagues (2019) found that NAC decreased extinction responding and reduced reinstatement of nicotine seeking. However, this effect was only observed with chronic NAC treatment [359].
Human Studies:
Clinical research has yet to follow up on the promising pre-clinical findings with ceftriaxone. However, there has been clinical exploration into NAC as a smoking cessation pharmacotherapy. In an open-label pilot trial of NAC in adult cigarette smokers (N=19), 4 weeks of NAC treatment (1200 mg, twice daily), in combination with varenicline (1 mg, twice daily), reduced cigarettes smoked per day [360]. A more recent randomized controlled trial has also been conducted among non-treatment seeking smokers (N=16). In this study, NAC treatment (1200 mg, twice daily) significantly decreased nicotine craving and positive affect during smoking abstinence [361]. In another trial, NAC treatment (1200 mg-3000 mg/day for 24 weeks) also helped reduce smoking among nicotine-dependent pathological gamblers (N=28] [362]. In contrast to these positive findings, Knackstedt and Colleagues (2009, N=33) reported that NAC (2400 mg/day, 4-weeks) treatment had no effect on daily cigarette use, CO levels, cigarette craving, or nicotine withdrawal [363].
The microglial inhibiting effects of pioglitazone have also been found to reduce nicotine craving [364]. In this trial, cigarette smokers, not interested in smoking cessation, were randomized to either active (45 mg, N=14) or placebo (0 mg, N=13) pioglitazone maintenance. During cue-exposure sessions, pioglitazone reduced nicotine craving but did not significantly alter cigarette self-administration or positive subjective response to smoking. Similarly, minocycline (200 mg/day, 4 days) has been shown to reduce craving for cigarettes but without altering responses to nicotine in non-treatment seeking smokers (N= 12) [365].
3.8. Cannabis Effects on Glia
There are robust data to suggest that the endocannabinoid system is sensitive to changes in glial activity. Acute exposure to cannabinoids [e.g., delta-9-tetrahydrocannabinol (THC), cannabidiol, and cannabinol] inhibits the production of inflammatory cytokines and chemokines [366–369]. However, chronic exposure may have different immunomodulatory effects. Individuals with cannabis use disorder show increases in plasma pro-inflammatory cytokines (IL-1β, IL-6, IL-8, and TNF-α) compared to individuals with no history of cannabis use [370].
The data suggest that exogenous cannabinoids interact with glia through the cannabinoid CB2 receptors. Upon activation of microglia, there is a significant increase in the expression of CB2 receptors [371–373]. In vitro, microglia produce endocannabinoids at higher levels than neurons [373], suggesting that endocannabinoid production by activated microglial cells could play a role in the pathophysiology of cannabis use disorder. Of particular interest has been the involvement of cannabinoid-glia interactions in THC-induced cognitive impairment. Studies have implicated the involvement of microglia in learning and motor coordination deficits induced by THC administration [374, 375]. However, in rodent models of THC-induced cognitive impairment, the CB1 receptor has been shown to be of particular importance. THC-induced deficits in learning and motor coordination were not observed in homozygous CB1 knockout mice [374].
Few studies have examined the effects of exogenous cannabinoids on other types of glia. Nonetheless, THC treatment was shown to increase the levels of the astrocyte marker GFAP in rats. Region-specific alternations in astrocyte reactivity have also been observed [376–377].
Pre-clinical Studies:
The author could find no studies examining the interaction between glial modulators and the rewarding effects of cannabis. Reliable, robust rewarding effects of cannabinoids are difficult to observe in pre-clinical models [378]. Impairment in cognitive, executive and emotional processes are some of the more severe outcomes related to heavy cannabis use [379]. In this treatment capacity, glial modulators have shown potential clinical utility. Minocycline prevented the development of THC-induced cognitive deficits [374]. Similarly, ibudilast blocked the cognitive effects of THC [375], which contribute to the adverse effects of chronic cannabis use [335, 380].
Human Studies:
It appears as though only NAC has been tested in human subjects. First, an open-label pilot trial found that NAC (1200 mg, twice daily) significantly decreased self-reported marijuana use and craving among young marijuana users (N=24) [381]. The investigators followed this study with a randomized controlled trial using the same dosing regimen among treatment-seeking cannabis-dependent adolescents (N=116). Active NAC treatment more than doubled the odds of having a negative cannabinoid urine toxicology result during the 8-week study [382]. The most recent trial of NAC was a 12-week double-blind randomized placebo-controlled investigation among treatment-seeking adults with cannabis use disorder (N=302), treated with 1200 mg of NAC, twice per day. However, this study found that NAC treatment did not alter abstinence rates [383]. Table 4 summarizes the pre-clinical and clinical findings of the effects of glial modulators on the abuse potential of nicotine and cannabis.
Table 4.
Effects of Glial Modulators on the Abuse Potential of Nicotine and Cannabis
| Preclinical Studies | |||||
|---|---|---|---|---|---|
| Glial Modulator | Mechanism of Action | Dose | Behavioral Outcome | Result | Citation |
| Nicotine | |||||
| Ceftriaxone | Cephalosporin Antibiotic & Upregulator of Glutamate Transporter-1 (GLT-1) and Cystine/Glutamate Antiporter (xCT) | 200 mg/kg | Rewarding Effects | -- | Alajaji et al., 2013 |
| Withdrawal | ↓ | ||||
| Reinstatement of Drug Seeking | ↓ | ||||
| 200 mg/kg | Reinforcing Effects | ↓ | Sari et al., 2016 | ||
| 200 mg/kg | Rewarding Effects | ↓ | Philogene-Khalid et al., 2017 | ||
| N-Acetylcysteine | Upregulator of GLT-1 & Glial Cystine-glutamate Exchange Activator | 15, 30 & 120 mg/kg, | Rewarding Effects | ↓ | Bowers et al., 2016 |
| 15, 30 & 120 mg/kg | Withdrawal | ↓ | |||
| 100 mg/kg | Reinstatement of Drug Seeking | ↓ | Gipson et al., 2013 | ||
| 60 mg/kg | Reinstatement of Drug Seeking | ↓ | Ramirez-Niño & D’Souza 2013 | ||
| 60 mg/kg | Reinforcing Effects | ↓ | |||
| 100 mg/kg | Reinstatement of Drug Seeking | ↓ | Goenaga et al., 2019 | ||
| 100 mg/kg | Withdrawal | ↓ | |||
| 100 mg/kg | Reinstatement of Drug Seeking | ↓ | Moro et al., 2019 | ||
| 100 mg/kg | Reinstatement of Drug Seeking | ↓ | Namba et al., 2019 | ||
| 100 mg/kg | Reinstatement of Drug Seeking | ↓ | Powel et al., 2019 | ||
| Cannabis | |||||
| Minocycline | Tetracycline Antibiotic & Microglial Inhibitor | 40 mg/kg | Cognitive Deficits | ↓ | Cutando et al., 2013 |
| Ibudilast | Phosphodiesterase (PDE) & Tumor Necrosis Factor-Alpha (TNF-α) Inhibitor | 3.5 mg/kg | Cognitive Deficits | ↓ | Zamberletti et al., 2015 |
| Clinical Studies | |||||
| Glial Modulator | Mechanism of Action | Dose | Behavioral Outcome | Result | Citation |
| Nicotine | |||||
| Minocycline | Tetracycline Antibiotic & Microglial Inhibitor | 200 mg | Craving | ↓ | Sofuoglu et al., 2009 |
| Pioglitazone | Peroxisome Proliferator-Activated Receptor-Gamma Activator & Cytokine Inhibitor | 45 mg | Craving | ↓ | Jones et al., 2017 |
| N-Acetylcysteine | Upregulator of GLT-1 & Glial Cystine-glutamate Exchange Activator | 2400 mg | Reinforing Effects, Craving, Abstinence, Withdrawal | -- | Knackstedt 2009 |
| 3600 mg | Craving | ↓ | Schmaal et al., 2011 | ||
| 1200 – 3000 mg | Abstinence | ↑ | Grant et al., 2014 | ||
| 2400 mg | Abstinence | ↑ | McClure et al., 2015 | ||
| 2400 mg | Craving | ↓ | Froeliger et al., 2015 | ||
| Cannabis | |||||
| N-Acetylcysteine | Upregulator of GLT-1 & Glial Cystine-glutamate Exchange Activator | 1200 mg | Craving | ↓ | Gray et al., 2010 |
| 1200 mg | Abstinence | ↑ | Gray et al., 2012 | ||
| 1200 mg | Abstinence | -- | Gray et al., 2017 | ||
4.0. Conclusions
Neuroimmune signaling is emerging as a significant contributor to the development and maintenance of drug and alcohol abuse. Though we are just beginning to understand how, studies suggest that activation and long-term modulation of glial function contributes to the rewarding properties of addictive drugs, sensitization to drug effects, withdrawal, and the loss of behavioral control that is characteristic of these disorders. There remains much to learn concerning how glia and neuroinflammatory responses contribute to the pathological features of SUDs. The acute vs chronic effects of drugs of abuse have yet to be conclusively determined. As such, it will be important to distinguish the immediate impact of drug-glial interaction on acute drug pharmacodynamics from the long-term effects that may have a persistent impact on brain function. Additionally, most of the studies for the role of glial in drug abuse focus on astrocytes and microglia, leaving the role of other glia types undefined. Furthermore, pre-clinical research suggests that there may be significant sexual dimorphism in how psychoactive drugs interact with glia. Because of the exploratory nature of most human studies, the influence of sex is rarely considered.
Pre-clinical studies employing anti-inflammatory drugs, or medications that modulate glia in other ways, have shown effectiveness in models of drug reward, reinforcement, and relapse. Findings of the therapeutic utility of medications that alter glutamate function (via glial mechanisms) have been some of the most consistently observed across drugs of abuse. Many of these medication findings have even been replicated in independent samples. Greater standardization of methodology across pre-clinical studies, and faster pace of research, make it easier to draw conclusions of treatment efficacy. The body of literature on clinical studies of glial modulators are more fragmented, with the same medication rarely being tested under the same experimental conditions.
Like, preclinical studies, clinical studies have yielded promising results, though not as robust. The most common finding among clinical trials has been medication effects on craving. Significant attenuation of the direct pharmacological effects of drugs of abuse is rare in clinical trials and may be a high bar for this class of medications. However, this should not lessen enthusiasm for continued clinical study, as craving is a significant contributor to the chronic relapsing nature of drug abuse. However, before we place limitations on the potential of glial modulators, we should note that clinical trials are often limited by dosing and maintenance conditions with established safety and efficacy for current FDA-approved indications. These parameters may not be applicable to the drug’s effects on glia, making it difficult to determine if a robust effect on glial was achieved. In fact, most clinical trials do not assess for changes in target glial activity, making proof-of-concept of the treatment mechanism difficult to determine.
In sum, each class of addictive drug appears to interact with glia in unique and multiple ways. We have just begun to understand the role of the complex interplay between the two systems in the development and maintenance of SUDs. This interaction is further complicated by variability in the neuronal mechanisms across the drugs classes and the resulting differences in patterns of use. A better understanding of the relationship between glia and the pathophysiology of addiction should allow for more precise clinical exploration of glial-directed treatments for SUDs.
Table 2.
Effects of Glial Modulators on the Abuse Potential of Psychostimulants
| Preclinical Studies | |||||
|---|---|---|---|---|---|
| Glial Modulator | Mechanism of Action | Dose | Behavioral Outcome | Result | Citation |
| N-Acetylcysteine | Glutamate Transporter-1 (GLT-1) Upregulator & Glial Cystine-glutamate Exchange Activator | 60 mg/kg | Reinstatement of Drug Seeking | ↓ | Baker et al., 2003 |
| 60 mg/kg | Reinstatement of Drug Seeking | ↓ | Madayag et al., 2007 | ||
| 100 mg/kg | Reinstatement of Drug Seeking | ↓ | Moussawi et al., 2011 | ||
| 100 mg/kg | Reinstatement of Drug Seeking | ↓ | Reichel et al., 2011 | ||
| 90 mg/kg | Reinstatement of Drug Seeking | ↓ | Murray et al., 2012 | ||
| 90 mg/kg | Reinforcing Effects | -- | |||
| 100 mg/kg | Reinstatement of Drug Seeking | ↓ | Reissner et al., 2015 | ||
| 30, 60, & 120 mg/kg | Reinforcing Effects & Reinstatement of Drug Seeking | -- | Charntikov et al., 2018 | ||
| Propentofylline | Phosphodiesterase (PDE) inhibitor & GLT-1 Upregulator | 3 μM | Reinstatement of Drug Seeking | ↓ | Narita et al., 2006 |
| 10 mg/kg | Reinstatement of Drug Seeking | ↓ | Reissner et al., 2014 | ||
| 100 & 200 mg/kg | Reinstatement of Drug Seeking | ↓ | Sari et al., 2009 | ||
| 200 mg/kg | Reinstatement of Drug Seeking | ↓ | Knackstedt et al., 2010 | ||
| 200 mg/kg | Reinforcing Effects | ↓ | Ward et al., 2011 | ||
| 200 mg/kg | Rewarding Effects | -- | Abulseoud et al., 2012 | ||
| 200 mg/kg | Reinforcing Effects | ↓ | |||
| Ibudilast | Phosphodiesterase & Tumor Necrosis Factor-Alpha (TNF-α) Inhibitor | 7.5 mg/kg | Reinstatement of Drug Seeking | ↓ | Beardsley et al., 2010 |
| 7.5 & 10 mg/kg | Reinforcing Effects | ↓ | Snider et al., 2013 | ||
| 7.5 & 10 mg/kg | Rewarding Effects | ↓ | Poland et al., 2016 | ||
| Minocycline | Tetracycline Antibiotic & Microglial Inhibitor | 60vmg/kg | Reinforcing Effects | ↓ | Snider et al., 2013 |
| 50 mg/kg | Reinforcing Effects | ↓ | Northcutt et al., 2015 | ||
| 40 mg/kg | Reinforcing Effects | ↓ | Attarzadeh-Yazdi et al., 2014 | ||
| 40 mg/kg | Reinstatement of Drug Seeking | ↓ | |||
| Pioglitazone | Peroxisome Proliferator-Activated Receptor-Gamma (PPARγ) & Cytokine Inhibitor | 50 mg/kg | Reinstatement of Drug Seeking | ↓ | Miller et al., 2016 |
| (+)-Naltrexone | Non-Opioid Isomer of the Opioid Receptor Antagonist & Toll-Like Receptor-4 (TLR-4) Antagonist | 30 mg/kg | Reinstatement of Drug Seeking | -- | Theberge et al., 2013 |
| (+)-Naloxone | Non-Opioid Isomer of the Opioid Receptor Antagonist & TLR-4 Antagonist | 5 mg/kg | Rewarding Effects | ↓ | Northcutt et al., 2015 |
| 5 mg/kg | Reinforcing Effects | ↓ | |||
| (+)-Naloxone (+)-Naltrexone | Non-Opioid Isomer of the Opioid Receptor Antagonist & TLR-4 Antagonist | 10, 17, 32 & 50 mg/kg | Reinforcing Effects | -- | Tanda et al., 2016 |
| MS-153 | Glial Glutamate Transport Activator | 10mg/kg | Rewarding Effects | ↓ | Nakagawa et al., 2005 |
| Clinical Studies | |||||
| Glial Modulator | Mechanism of Action | Dose | Behavioral Outcome | Result | Citation |
| Pentoxifylline | Cytokine inhibitor | 1200 mg | Abstinence | -- | Ciraulo et al., 2005 |
| Minocycline | Tetracycline Antibiotic & Microglial Inhibitor | 200 mg | Positive Subjective Effects | ↓ | Sofuogo et al., 2011 |
| 200 mg | Reinforcing Effects | -- | |||
| Celecoxib | Nonsteroidal Anti-Inflammatory (Cyclooxygenase-2 Inhibitor) | 200 mg | Abstinence & Craving | -- | Reid et al., 2005 |
| N-Acetylcysteine | Upregulator of GLT-1 & Glial Cystine-glutamate Exchange Activator | 600 mg | Craving | ↓ | LaRowe et al., 2007 |
| 2400 & 3600 mg | Abstinence | ↓ | Mardikian et al., 2007 | ||
| 1200-2400 mg | Positive Subjective Effects | -- | Amen et al., 2011 | ||
| 1200-2400 mg | Craving | ↓ | |||
| 1200 mg | Craving | ↓ | Mousavi et al., 2015 | ||
| 2400 mg | Craving | ↓ | Back et al., 2016 | ||
| 2400 mg | Craving & Cue-Reactivity | ↓ | Levi-Bolin et al., 2017 | ||
| 2400 mg | Reinforcing Effects | -- | |||
| Pioglitazone | PPARγ Activator & Cytokine Inhibitor | 45 mg | Reinforcing Effects | ↓ | Schmitz et al., 2017 |
| 45 mg | Craving | ↓ | |||
Key Points.
Pre-clinical findings show that drugs of abuse have significant modulatory actions upon neuroinflammatory signaling, the most investigated of which are their interaction with microglia and astrocytes.
There is evidence that the interaction between drugs of abuse and glia contributes to the development, maintenance and course of substance use disorders.
Though clinical studies of glial modulators as treatments of substance abuse have not yielded the same robust findings as pre-clinical models, there are sufficient positive results to justify continued investigation.
Acknowledgments
Funding: Financial support for the preparation of this manuscript was provided by the National Institute on Drug Abuse (Baltimore, MD) grants R21DA043199 and R21DA040225 to the author.
Footnotes
Compliance with Ethical Standards
Conflicts of Interest: Over the past three years, Dr. Jones has received partial salary support for a clinical trial partly funded by Cerecor Inc. Dr. Jones is also the recipient of an investigator-initiated grant from Merck Pharmaceuticals studying HIV among methamphetamine users.
References
- 1.Koob GF, Volkow ND. Neurobiology of addiction: a neurocircuitry analysis. Lancet Psychiatry. 2016;3(8):760–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.National Institute on Drug Abuse. The science of drug use and addiction: the basics. 2014. http://www.drugabuse.gov/sites/default/files/mediaguide_web_3_0.pdf. Accessed 15 Dec 2019.
- 3.UNODC. World drug report. 2018. UN, New York. 10.18356/d29e3f27-en. Accessed 17 Nov 2019. [DOI] [Google Scholar]
- 4.Mattick RP, Kimber J, Breen C, Davoli M. Buprenorphine maintenance versus placebo or methadone maintenance for opioid dependence. Cochrane Database Syst Rev. 2004;(3):CD002207. [DOI] [PubMed] [Google Scholar]
- 5.Sordo L, Barrio G, Bravo MJ, Indave BI, Degenhardt L, Wiessing L, et al. Mortality risk during and after opioid substitution treatment: systematic review and meta-analysis of cohort studies. BMJ. 2017;357:j1550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Timko C, Schultz NR, Cucciare MA, Vittorio L, Garrison-Diehn C. Retention in medication-assisted treatment for opiate dependence: a systematic review. J Addict Dis. 2016;35(1):22–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.World Health Organization. Management of substance abuse. 2018. https://www.who.int/substance_abuse/facts/en/. Accessed 23 Oct 2019.
- 8.Klein JW. Pharmacotherapy for substance use disorders. Med Clin N Am. 2016;100(4):891–910. [DOI] [PubMed] [Google Scholar]
- 9.Rösner S, Hackl‐Herrwerth A, Leucht S, Vecchi S, Srisurapanont M, Soyka M. Opioid antagonists for alcohol dependence. Cochrane Database Syst Rev 2010;(12):CD001867. [DOI] [PubMed] [Google Scholar]
- 10.Jonas DE, Amick HR, Feltner C, Bobashev G, Thomas K, Wines R, et al. Pharmacotherapy for adults with alcohol use disorders in outpatient settings: a systematic review and metaanalysis pharmacotherapy for alcohol use disorders. JAMA. 2014;311(18):1889–900. [DOI] [PubMed] [Google Scholar]
- 11.Aubin H-J, Luquiens A, Berlin I. Pharmacotherapy for smoking cessation: pharmacological principles and clinical practice. Br J Clin Pharmacol. 2014;77(2):324–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Jain R, Majumder P, Gupta T. Pharmacological intervention of nicotine dependence. Biomed Res Int. 2013;2013:278392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Henningfield JE, Fant RV, Buchhalter AR, Stitzer ML. Pharmacotherapy for nicotine dependence. CA Cancer J Clin. 2005;55(5):281–99. [DOI] [PubMed] [Google Scholar]
- 14.Mathers CD, Loncar D. Projections of global mortality and burden of disease from 2002 to 2030. PLoS Med. 2006;3(11):e442. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Choi NG, DiNitto DM, Marti CN. Marijuana use among adults: Initiation, return to use, and continued use versus quitting over a one-year follow-up period. Drug Alcohol Depend. 2018;182:19–26. [DOI] [PubMed] [Google Scholar]
- 16.Hasin DS, Saha TD, Kerridge BT, Goldstein RB, Chou SP, Zhang H, et al. Prevalence of marijuana use disorders in the United States between 2001–2002 and 2012–2013 prevalence of marijuana use disorders in the United States prevalence of marijuana use disorders in the United States. JAMA Psychiatry. 2015;72(12):1235–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Ransom BR, Sontheimer H. The neurophysiology of glial cells. J Clin Neurophysiol. 1992;9(2):224–51. [DOI] [PubMed] [Google Scholar]
- 18.Araque A, Carmignoto G, Haydon Philip G, Oliet Stéphane HR, Robitaille R, Volterra A. Gliotransmitters travel in time and space. Neuron. 2014;81(4):728–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Bilbo SD, Schwarz JM. The immune system and developmental programming of brain and behavior. Front Neuroendocrinol. 2012;33(3):267–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Chung W-S, Welsh CA, Barres BA, Stevens B. Do glia drive synaptic and cognitive impairment in disease? Nat Neurosci. 2015;18(11):1539–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Azevedo FAC, Carvalho LRB, Grinberg LT, Farfel JM, Ferretti REL, Leite REP, et al. Equal numbers of neuronal and nonneuronal cells make the human brain an isometrically scaled-up primate brain. J Comp Neurol. 2009;513(5):532–41. [DOI] [PubMed] [Google Scholar]
- 22.Herculano-Houzel S. The glia/neuron ratio: how it varies uniformly across brain structures and species and what that means for brain physiology and evolution. Glia. 2014;62(9):1377–91. [DOI] [PubMed] [Google Scholar]
- 23.Pelvig DP, Pakkenberg H, Stark AK, Pakkenberg B. Neocortical glial cell numbers in human brains. Neurobiol Aging. 2008;29(11):1754–62. [DOI] [PubMed] [Google Scholar]
- 24.Sofroniew MV, Vinters HV. Astrocytes: biology and pathology. Acta Neuropathol. 2010;119(1):7–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Butt AM. Macroglial lineages. In: Square LR (eds) Encyclopedia of Neuroscience. Academic Press, Cambridge, Massachusetts; 2009. [Google Scholar]
- 26.Fetler L, Amigorena S. Brain under surveillance: the microglia patrol. Science. 2005;309(5733):392–3. [DOI] [PubMed] [Google Scholar]
- 27.Eric Thomas W. Brain macrophages: evaluation of microglia and their functions. Brain Res Rev. 1992;17(1):61–74. [DOI] [PubMed] [Google Scholar]
- 28.Nimmerjahn A, Kirchhoff F, Helmchen F. Resting microglial cells are highly dynamic surveillants of brain parenchyma in vivo. Science. 2005;308(5726):1314–8. [DOI] [PubMed] [Google Scholar]
- 29.Boucsein C, Zacharias R, Färber K, Pavlovic S, Hanisch U-K, Kettenmann H. Purinergic receptors on microglial cells: functional expression in acute brain slices and modulation of microglial activation in vitro. Eur J Neurosci. 2003;17(11):2267–76. [DOI] [PubMed] [Google Scholar]
- 30.Taylor DL, Diemel LT, Pocock JM. Activation of microglial group III metabotropic glutamate receptors protects neurons against microglial neurotoxicity. J Neurosci. 2003;23(6):2150–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Kimelberg HK, Norenberg MD. Astrocytes. Sci Am. 1989;260(4):66–72. [DOI] [PubMed] [Google Scholar]
- 32.Schousboe A, Sarup A, Bak LK, Waagepetersen HS, Lars-son OM. Role of astrocytic transport processes in glutamatergic and GABAergic neurotransmission. Neurochem Int. 2004;45(4):521–7. [DOI] [PubMed] [Google Scholar]
- 33.Magistretti PJ, Pellerin L. Astrocytes couple synaptic activity to glucose utilization in the brain. News Physiol Sci. 1999;14:177–82. [DOI] [PubMed] [Google Scholar]
- 34.Silver J, Miller JH. Regeneration beyond the glial scar. Nat Rev Neurosci. 2004;5(2):146–56. [DOI] [PubMed] [Google Scholar]
- 35.Edgar JM, Garbern J. The myelinated axon is dependent on the myelinating cell for support and maintenance: molecules involved. J Neurosci Res. 2004;76(5):593–8. [DOI] [PubMed] [Google Scholar]
- 36.Dai X, Lercher LD, Clinton PM, Du Y, Livingston DL, Vieira C, et al. The trophic role of oligodendrocytes in the basal forebrain. J Neurosci. 2003;23(13):5846–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Del Bigio MR. The ependyma: a protective barrier between brain and cerebrospinal fluid. Glia. 1995;14:1–13. [DOI] [PubMed] [Google Scholar]
- 38.Barry DS, Pakan JMP, McDermott KW. Radial glial cells: key organisers in CNS development. Int J Biochem Cell Biol. 2014;46:76–9. [DOI] [PubMed] [Google Scholar]
- 39.Malatesta P, Appolloni I, Calzolari F. Radial glia and neural stem cells. Cell Tissue Res. 2008;331(1):165–78. [DOI] [PubMed] [Google Scholar]
- 40.Hyman SE, Malenka RC, Nestler EJ. Neural mechanisms of addiction: the role of reward-related learning and memory. Ann Rev Neurosci. 2006;29(1):565–98. [DOI] [PubMed] [Google Scholar]
- 41.Kalivas PW. The glutamate homeostasis hypothesis of addiction. Nat Rev Neurosci. 2009;10:561. [DOI] [PubMed] [Google Scholar]
- 42.Kauer JA, Malenka RC. Synaptic plasticity and addiction. Nat Rev Neurosci. 2007;8:844. [DOI] [PubMed] [Google Scholar]
- 43.Volkow ND, Wise RA, Baler R. The dopamine motive system: implications for drug and food addiction. Nat Rev Neurosci. 2017;18:741. [DOI] [PubMed] [Google Scholar]
- 44.Koob GF. Neural mechanisms of drug reinforcement. Anne NY Acad Sci. 1992;654(1):171–91. [DOI] [PubMed] [Google Scholar]
- 45.Lacagnina MJ, Rivera PD, Bilbo SD. Glial and neuroimmune mechanisms as critical modulators of drug use and abuse. Neuropsychopharmacology. 2016;42:156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Linker KE, Cross SJ, Leslie FM. Glial mechanisms underlying substance use disorders. Eur J Neurosci. 2019;50(3):2574–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Zhang Q, Haydon PG. Roles for gliotransmission in the nervous system. J Neural Transm (Vienna). 2005;112(1):121–5. [DOI] [PubMed] [Google Scholar]
- 48.Jones JD, Comer SD. A review of human drug self-administration procedures. Behav Pharmacol. 2013;24(5–6):384–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Bidlack JM. Detection and function of opioid receptors on cells from the immune system. Clin Diagn Lab Immunol. 2000;7(5):719–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Gomez-Flores R, Weber RJ. Opioids, opioid receptors, and the immune system. In: Plotnikoff NP, Faith RE, Murgo AJ, Good RA, editors. Cytokines, stress and immunity. Boca Raton: CRC Press; 1998. p. 281–314. [Google Scholar]
- 51.Sibinga NE, Goldstein A. Opioid peptides and opioid receptors in cells of the immune system. Annu Rev Immunol. 1988;6:219–49. [DOI] [PubMed] [Google Scholar]
- 52.Cheng PY, Liu-Chen LY, Pickel VM. Dual ultrastructural immunocytochemical labeling of mu and delta opioid receptors in the superficial layers of the rat cervical spinal cord. Brain Res. 1997;778:367–80. [DOI] [PubMed] [Google Scholar]
- 53.Commons KG, Milner TA. Cellular and subcellular localization of delta opioid receptor immunoreactivity in the rat dentate gyrus. Brain Res. 1996;738:181–95. [DOI] [PubMed] [Google Scholar]
- 54.Drake CT, Chang PC, Harris JA, Milner TA. Neurons with mu opioid receptors interact indirectly with enkephalin-containing neurons in the rat dentate gyrus. Exp Neurol. 2002;176:254–61. [DOI] [PubMed] [Google Scholar]
- 55.Svingos AL, Moriwaki A, Wang JB, Uhl GR, Pickel VM. Ultrastructural immunocytochemical localization of mu-opioid receptors in rat nucleus accumbens: extrasynaptic plasmalemmal distribution and association with Leu5-enkephalin. J Neurosci. 1996;16:4162–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Harris JA, Chang PC, Drake CT. Kappa opioid receptors in rat spinal cord: sex-linked distribution differences. Neuroscience. 2004;124:879–90. [DOI] [PubMed] [Google Scholar]
- 57.Svingos AL, Colago EE. Kappa-opioid and NMDA glutamate receptors are differentially targeted within rat medial prefrontal cortex. Brain Res. 2002;946:262–71. [DOI] [PubMed] [Google Scholar]
- 58.Ubink R, Calza L, Hökfelt T. ‘Neuro’-peptides in glia: focus on NPY and galanin. Trends Neurosci. 2003;26(11):604–9. [DOI] [PubMed] [Google Scholar]
- 59.Vallejo R, de Leon-Casasola O, Benyamin R. Opioid therapy and immunosuppression: a review. Am J Ther. 2004;11(5):354–65. [DOI] [PubMed] [Google Scholar]
- 60.Liang X, Liu R, Chen C, Ji F, Li T. Opioid system modulates the immune function: a review. Transl Perioper Pain Med. 2016;1(1):5–13. [PMC free article] [PubMed] [Google Scholar]
- 61.Mellon RD, Bayer BM. Evidence for central opioid receptors in the immunomodulatory effects of morphine: review of potential mechanism(s) of action. J Neuroimmunol. 1998;83:19–28. [DOI] [PubMed] [Google Scholar]
- 62.Pathan H, Williams J. Basic opioid pharmacology: an update. Br J Pain. 2012;6(1):11–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Beitner-Johnson D, Guitart X, Nestler EJ. Glial fibrillary acidic protein and the mesolimbic dopamine system: regulation by chronic morphine and Lewis–Fischer strain differences in the rat ventral tegmental area. J Neurochem. 1993;61:1766–73. [DOI] [PubMed] [Google Scholar]
- 64.Hol EM, Pekny M. Glial fibrillary acidic protein (GFAP) and the astrocyte intermediate filament system in diseases of the central nervous system. Curr Opin Cell Biol. 2015;32:121–30. [DOI] [PubMed] [Google Scholar]
- 65.Hutchinson MR, Lewis SS, Coats BD, Skyba DA, Crysdale NY, Berkelhammer DL, et al. Reduction of opioid withdrawal and potentiation of acute opioid analgesia by systemic AV411 (ibudilast). Brain Behav Immun. 2009;23(2):240–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Hutchinson MR, Zhang Y, Shridhar M, Evans JH, Buchanan MM, Zhao TX, et al. Evidence that opioids may have toll-like receptor 4 and MD-2 effects. Brain Behav Immun. 2010;24(1):83–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Schwarz JM, Hutchinson MR, Bilbo SD. Early-life experience decreases drug-induced reinstatement of morphine CPP in adulthood via microglial-specific epigenetic programming of antiinflammatory IL-10 expression. J Neurosci. 2011;31:17835–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Borish LC, Steinke JW. 2. Cytokines and chemokines. J Allergy Clin Immunol. 2003;111(2 Suppl. 2):S460–75. [DOI] [PubMed] [Google Scholar]
- 69.El-Hage N, Gurwell JA, Singh IN, Knapp PE, Nath A, Hauser KF. Synergistic in-creases in intracellular C a2+, and the release of MCP-1, RANTES, and IL-6 by astrocytes treated with opiates and HIV-1 Tat. Glia. 2005;50:91–106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.El-Hage N, Wu G, Wang J, Ambati J, Knapp PE, Reed JL, et al. HIV-1 Tat and opiate-induced changes in astrocytes promote chemotaxis of microglia through the expression of MCP-1 and alternative chemokines. Glia. 2006;53:132–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Niwa M, Nitta A, Yamada Y, Nakajima A, Saito K, Seishima M, et al. Tumor necrosis factor-alpha and its inducer inhibit morphine-induced re-warding effects and sensitization. Biol Psychiatry. 2007;62:658–68. [DOI] [PubMed] [Google Scholar]
- 72.Sawaya BE, Deshmane SL, Mukerjee R, Fan S, Khalili K. TNF alpha production in morphine-treated human neural cells is NFkappa B-dependent. J Neuroimmune Pharmacol. 2009;4:140–9. [DOI] [PubMed] [Google Scholar]
- 73.Hu S, Sheng WS, Lokensgard JR, Peterson PK. Morphine induces apoptosis of human microglia and neurons. Neuropharmacology. 2002;42(6):829–36. [DOI] [PubMed] [Google Scholar]
- 74.Horvath RJ, DeLeo JA. Morphine enhances microglial migration through modulation of P2X4 receptor signaling. J Neurosci. 2009;29(4):998–1005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Li Y, Li H, Zhang Y, Sun X, Hanley GA, LeSage G, et al. Tolllike receptor 2 is required for opioids-induced neuronal apoptosis. Biochem Biophys Res Commun. 2010;391(1):426–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.He L, Li H, Chen L, Miao J, Jiang Y, Zhang Y, et al. Toll-like receptor 9 is required for opioid-induced microglia apoptosis. PLoS One. 2011;6(4):e18190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Medzhitov R, Preston-Hurlburt P, Janeway CA. A human homologue of the Drosophila Toll protein signals activation of adaptive immunity. Nature. 1997;388(6640):394–7. [DOI] [PubMed] [Google Scholar]
- 78.Jack CS, Arbour N, Manusow J, Montgrain V, Blain M, et al. TLR signaling tailors innate immune responses in human microglia and astrocytes. J Immunol. 2005;175:4320–30. [DOI] [PubMed] [Google Scholar]
- 79.Lehnardt S, Massillon L, Follett P, Jensen FE, Ratan R, et al. Activation of innate immunity in the CNS triggers neurodegeneration through a Toll-like receptor 4-dependent pathway. Proc Natl Acad Sci USA. 2003;100:8514–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Wang X, Loram LC, Ramos KM, de Jesus AJ, Thomas J, Reddy A, et al. Morphine activates neuroinflammation in a manner parallel to endotoxin. PNAS. 2012;109(16):6325–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Hutchinson MR, Lewis SS, Coats BD, Rezvani N, Zhang Y, Wieseler JL, et al. Possible involvement of toll-like receptor 4/ myeloid differentiation factor-2 activity of opioid inactive isomers causes spinal proinflammation and related behavioral consequences. Neuroscience. 2010;167:880–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Wise RA. Drug-activation of brain reward pathways. Drug Alcohol Depend. 1998;51(1–2):13–22. [DOI] [PubMed] [Google Scholar]
- 83.Nutt DJ, Lingford-Hughes A, Erritzoe D, Stokes PRA. The dopamine theory of addiction: 40 years of highs and lows. Nat Rev Neurosci. 2015;16:305. [DOI] [PubMed] [Google Scholar]
- 84.Wise RA, Bozarth MA. Brain substrates for reinforcement and drug self-administration. Prog Neuropsychopharmacol. 1981;5(5):467–74. [DOI] [PubMed] [Google Scholar]
- 85.Suzuki T, Masukawa Y, Shiozaki Y, Misawa M. Potentiation of pentazocine conditioned place preference by tripelennamine in rats. Psychopharmacology (Berl). 1991;105(1):9–12. [DOI] [PubMed] [Google Scholar]
- 86.Bland ST, Hutchinson MR, Maier SF, et al. The glial activation inhibitor AV411 reduces morphine-induced nucleus accumbens dopamine release. Brain Behav Immun. 2009;23:492–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Hutchinson MR, Bland ST, Johnson KW, et al. Opioid-induced glial activation: mechanisms of activation and implications for opioid analgesia, dependence and reward. Sci World J. 2007;7:98–111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Hutchinson MR, Northcutt A, Hiranita T, Wang X, Lewis SS, Thomas J, et al. Opioid activation of toll-like receptor 4 contributes to drug reinforcement. J Neurosci. 2012;32:11187–200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Theberge FR, Li X, Kambhampati S, Pickens CL, St Laurent R, Bossert JM, et al. Effect of chronic delivery of the Toll-like receptor 4 antagonist (+)-naltrexone on incubation of heroin craving. Biol Psychiatry. 2013;73(8):729–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Tanda G, Mereu M, Hiranita T, Quarterman JC, Coggiano M, Katz JL. Lack of specific involvement of (+)-naloxone and (+)-naltrexone on the reinforcing and neurochemical effects of cocaine and opioids. Neuropsychopharmacol. 2016;41(11):2772–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.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(1–2):203–12. [DOI] [PubMed] [Google Scholar]
- 92.Ledeboer A, Hutchinson MR, Watkins LR, Johnson KW. Ibudilast (AV-411): a new class therapeutic candidate for neuropathic pain and opioid withdrawal syndromes. Expert Opin Investig Drugs. 2007;16:935–50. [DOI] [PubMed] [Google Scholar]
- 93.Dugosh K, Abraham A, Seymour B, McLoyd K, Chalk M, Festinger D. A systematic review on the use of psychosocial interventions in conjunction with medications for the treatment of opioid addiction. J Addict Med. 2016;10(2):93–103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Gregory E Norsted, Delaney A, AbdelMoaty S, Bas DB, Codeluppi S, Wigerblad G, et al. Pentoxifylline and propentofylline prevent proliferation and activation of the mammalian target of rapamycin and mitogen activated protein kinase in cultured spinal astrocytes. J Neurosci Res. 2013;91(2):300–12. [DOI] [PubMed] [Google Scholar]
- 95.Raghavendra V, Tanga FY, DeLeo JA. Attenuation of morphine tolerance, withdrawal-induced hyperalgesia, and associated spinal inflammatory immune responses by propentofylline in rats. Neuropsychopharmacology. 2004;29(2):327–34. [DOI] [PubMed] [Google Scholar]
- 96.Tawfik VL, Regan MR, Haenggeli C, Lacroix-Fralish ML, Nutile-McMenemy N, Perez N, et al. Propentofylline-induced astrocyte modulation leads to alterations in glial glutamate promoter activation following spinal nerve transection. Neuroscience. 2008;152:1086–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Narita M, Miyatake M, Narita M, Shibasaki M, Shindo K, Nakamura A, et al. Direct evidence of astrocytic modulation in the development of rewarding effects induced by drugs of abuse. Neuropsychopharmacology. 2006;31(11):2476–88. [DOI] [PubMed] [Google Scholar]
- 98.Hassel B, Paulsen RE, Johnsen A, Fonnum F. Selective inhibition of glial cell metabolism in vivo by fluorocitrate. Brain Res. 1992;576(1):120–4. [DOI] [PubMed] [Google Scholar]
- 99.Seyedaghamiri F, Heysieattalab S, Hosseinmardi N, Janahmadi M, Elahi-Mahani A, Salari F, et al. Hippocampal glial cells modulate morphine-induced behavioral responses. Physiol Behav. 2018;191:37–46. [DOI] [PubMed] [Google Scholar]
- 100.Cheng S, Hou J, Zhang C, Xu C, Wang L, Zou X, et al. Minocycline reduces neuroinflammation but does not ameliorate neuron loss in a mouse model of neurodegeneration. Sci Rep. 2015;5:10535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Arezoomandan R, Haghparast A. Administration of the glial cell modulator, minocycline, in the nucleus accumbens attenuated the maintenance and reinstatement of morphine-seeking behavior. Can J Physiol Pharmacol. 2016;94(3):257–64. 10.1139/cjpp-2015-0209 (Epub 2015 Aug 24). [DOI] [PubMed] [Google Scholar]
- 102.Kielian T, Drew PD. Effects of PPARy agonists on central nervous system inflammation. J Neurosci Res. 2003;71:315–25. [DOI] [PubMed] [Google Scholar]
- 103.de Guglielmo G, Kallupi M, Scuppa G, Stopponi S, Demopulos G, Gaitanaris G, et al. Analgesic tolerance to morphine is regulated by PPARγ. Br J Pharmacol. 2014;171(23):5407–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.de Guglielmo G, Melis M, De Luca MA, Kallupi M, Li HW, Niswender K, et al. PPARγ activation attenuates opioid consumption and modulates mesolimbic dopamine transmission. Neuropsychopharmacology. 2015;40(4):927–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.de Guglielmo G, Kallupi M, Scuppa G, Demopulos G, Gaitanaris G, Ciccocioppo R. Pioglitazone attenuates the opioid withdrawal and vulnerability to relapse to hero-in seeking in rodents. Psychopharmacology (Berl.). 2017;234(2):223–34. [DOI] [PubMed] [Google Scholar]
- 106.Ghavimi H, Hassanzadeh K, Maleki-Dizaji N, Azarfardian A, Ghasami S, Zolali E, et al. Pioglitazone prevents morphine antinociception tolerance and withdrawal symptoms in rats. Naunyn Schmiedebergs Arch Pharmacol. 2014;387(9):811–21. [DOI] [PubMed] [Google Scholar]
- 107.Grace PM, Maier SF, Watkins LR. Opioid-induced central immune signaling: implications for opioid analgesia. Headache. 2015;55(4):475–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Raghavendra V, Rutkowski MD, DeLeo JA. The role of spinal neuroimmune activation in morphine tolerance/hyperalgesia in neuropathic and sham-operated rats. J Neurosci. 2002;22(22):9980–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Watkins LR, Hutchinson MR, Johnston IN, Maier SF. Glia: novel counter-regulators of opioid analgesia. Trends Neurosci. 2005;28(12):661–9. [DOI] [PubMed] [Google Scholar]
- 110.Cui Y, Liao X-X, Liu W, Guo R-X, Wu Z-Z, Zhao C-M, et al. A novel role of minocycline: attenuating morphine antinociceptive tolerance by inhibition of p38 MAPK in the activated spinal microglia. Brain Behav Immun. 2008;22(1):114–23. [DOI] [PubMed] [Google Scholar]
- 111.Johnson KW, Matsuda K, Iwaki Y. Ibudilast for the treatment of drug addiction and other neurological conditions. Clin Invest. 2014;4:1–11. [Google Scholar]
- 112.Kosarmadar N, Ghasemzadeh Z, Rezayof A. Inhibition of microglia in the basolateral amygdala enhanced morphine-induced antinociception: possible role of GABAA receptors. Eur J Pharmacol. 2015;765:157–63. [DOI] [PubMed] [Google Scholar]
- 113.Mika J, Osikowicz M, Makuch W, Przewlocka B. Minocycline and pentoxifylline attenuate allodynia and hyperalgesia and potentiate the effects of morphine in rat and mouse models of neuropathic pain. Eur J Pharmacol. 2007;560:142–9. [DOI] [PubMed] [Google Scholar]
- 114.Raghavendra V, Tanga F, Rutkowski MD, DeLeo JA. Antihyperalgesic and morphine-sparing actions of propentofylline following peripheral nerve injury in rats: mechanistic implications of spinal glia and proinflammatory cytokines. Pain. 2003;104:655–64. [DOI] [PubMed] [Google Scholar]
- 115.Song P, Zhao ZQ. The involvement of glial cells in the development of morphine tolerance. Neurosci Res. 2001;39:281–6. [DOI] [PubMed] [Google Scholar]
- 116.Kim HP, Park H, Son KH, Chang HW, Kang SS. Biochemical pharmacology of bioflavonoids: implications for anti-inflammatory action. Arch Pharm Res. 2008;31(3):265–73. [DOI] [PubMed] [Google Scholar]
- 117.Cai Y, Kong H, Pan YB, Jiang L, Pan XX, Hu L, et al. Procyanidins alleviates morphine tolerance by inhibiting activation of NLRP3 inflammasome in microglia. J Neuroinflamm. 2016;13(1):53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.März P, Otten U, Miserez AR. Statins induce differentiation and cell death in neurons and astroglia. Glia. 2007;55(1):1–12. [DOI] [PubMed] [Google Scholar]
- 119.Sasaki S, Futagi Y, Ideno M, Kobayashi M, Narumi K, Furugen A, et al. Interaction of atorvastatin with the human glial transporter SLC16A1. Eur J Pharmacol. 2016;788:248–54. [DOI] [PubMed] [Google Scholar]
- 120.Yamamoto N, Fujii Y, Kasahara R, Tanida M, Ohora K, Ono Y, et al. Simvastatin and atorvastatin facilitates amyloid β-protein degradation in extracellular spaces by increasing neprilysin secretion from astrocytes through activation of MAPK/Erk1/2 pathways. Glia. 2016;64(6):952–62. [DOI] [PubMed] [Google Scholar]
- 121.Ghasemi F, Moradi A, Izadpanah E, Moloudi MR, Hassanzadeh K, Rahimmi A, et al. Simvastatin prevents morphine antinociceptive tolerance and withdrawal symptoms in rats. J Formos Med Assoc. 2015;114(5):399–406. [DOI] [PubMed] [Google Scholar]
- 122.Li Y, Shu Y, Ji Q, Liu J, He X, Li W. Attenuation of morphine analgesic tolerance by rosuvastatin in naïve and morphine tolerance rats. Inflammation. 2015;38(1):134–41. [DOI] [PubMed] [Google Scholar]
- 123.Li W, Li Y, Zhu S, Ji Q, Shu Y, Zhang L, Liu J. Rosuvastatin attenuated the existing morphine tolerance in rats with L5 spinal nerve transection through inhibiting activation of astrocytes and phosphorylation of ERK42/44. Neurosci Lett. 2015;584:314–9. [DOI] [PubMed] [Google Scholar]
- 124.Mansouri MT, Khodayar MJ, Tabatabaee A, Ghorbanzadeh B, Naghizadeh B. Modulation of morphine antinociceptive tolerance and physical dependence by co-administration of simvastatin. Pharmacol Biochem Behav. 2015;137:38–43. [DOI] [PubMed] [Google Scholar]
- 125.Pajohanfar NS, Mohebbi E, Hosseini-Bandegharaei A, Amin M, Vaseghi G, Amin B. Simvastatin prevents morphine-induced tolerance and dependence in mice. Biomed Pharmacother. 2017;93:406–11. [DOI] [PubMed] [Google Scholar]
- 126.Pajohanfar NS, Mohebbi E, Rad A, Pejhan A, Nazemi S, Amin B. Protective effects of atorvastatin against morphine-induced tolerance and dependence in mice. Brain Res. 2017;15(1657):333–9. [DOI] [PubMed] [Google Scholar]
- 127.Ozawa T, Nakagawa T, Shige K, Minami M, Satoh M. Changes in the expression of glial glutamate transporters in the rat brain accompanied with morphine dependence and naloxone-precipitated withdrawal. Brain Res. 2001;905(1):254–8. [DOI] [PubMed] [Google Scholar]
- 128.Nakagawa T, Fujio M, Ozawa T, Minami M, Satoh M. Effect of MS-153, a glutamate transporter activator, on the conditioned rewarding effects of morphine, methamphetamine and cocaine in mice. Behav Brain Res. 2005;156(2):233–9. [DOI] [PubMed] [Google Scholar]
- 129.Alshehri FS, Hakami AY, Althobaiti YS, Sari Y. Effects of ceftriaxone on hydrocodone seeking behavior and glial glutamate transporters in P rats. Behav Brain Res. 2018;347:368–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Shen H-W, Scofield MD, Boger H, Hensley M, Kalivas PW. Synaptic glutamate spillover due to impaired glutamate uptake mediates heroin relapse. J Neurosci. 2014;34(16):5649–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Zhou W, Kalivas PW. N-Acetylcysteine reduces extinction responding and induces enduring reductions in cue- and heroininduced drug-seeking. Biol Psychiatry. 2008;63(3):338–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Moran MM, McFarland K, Melendez RI, Kalivas PW, Seamans JK. Cystine/glutamate exchange regulates metabotropic glutamate receptor presynaptic inhibition of excitatory transmission and vulnerability to cocaine seeking. J Neurosci. 2005;25(27):6389–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Hodebourg R, Murray JE, Fouyssac M, Puaud M, Everitt BJ, Belin D. Heroin seeking becomes dependent on dorsal striatal dopaminergic mechanisms and can be decreased by N-acetylcysteine. Eur J Neurosci. 2019;50(3):2036–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Cooper ZD, Jones JD, Comer SD. Glial modulators: a novel pharmacological approach to altering the behavioral effects of abused substances. Expert Opin Investig Drugs. 2012;21(2):169–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Bachtella RK, Jones JD, Heinzerling CG, Beardsley PM, Comer SD. Glial and neuroinflammatory targets for treating substance use disorders. Drug Alcohol Depend. 2017;180:156–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Cooper ZD, Johnson KW, Pavlicova M, Glass A, Vosburg SK, Sullivan MA, et al. The effects of ibudilast, a glial activation inhibitor, on opioid withdrawal symptoms in opioid-dependent volunteers. Addict Biol. 2016;21:895–903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Cooper ZD, Johnson KW, Vosburg SK, Sullivan MA, Manubay J, Martinez D, et al. Effects of ibudilast on oxycodone-induced analgesia and subjective effects in opioid-dependent volunteers. Drug Alcohol Depend. 2017;1(178):340–7. 10.1016/j.drugalcdep.2017.04.029 (Epub 2017 Jun 17). [DOI] [PubMed] [Google Scholar]
- 138.Metz VE, Jones JD, Manubay J, Sullivan M, Mogali S, Segoshi A, et al. Effects of ibudilast on the subjective, reinforcing and analgesic effects of oxycodone in recently detoxified adults with opioid dependence. Neuropsychopharmacology. 2017;42(9):1825–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Mogali S, Jones JD, Manubay JM, Sullivan MS, Comer SD. Effects of minocycline on oxycodone-induced responses in humans. Drug Alcohol Depend. 2014. 10.1016/j.drugalcdep.2014.02.433. [DOI] [Google Scholar]
- 140.Arout CA, Waters AJ, MacLean RR, Compton P, Sofuoglu M. Minocycline does not affect experimental pain or addictionrelated outcomes in opioid maintained patients. Psychopharmacology. 2019;236(10):2857–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Jones JD, Bisaga A, Metz VE, Manubay JM, Mogali S, Ciccocioppo R, et al. The PPARγ agonist pioglitazone fails to alter the abuse potential of heroin, but does reduce heroin craving and anxiety. J Psychoact Drugs. 2018;50(5):390–401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Jones JD, Sullivan MA, Manubay JM, Mogali S, Metz VE, Ciccocioppo R, Comer SD. The effects of pioglitazone, a PPARγ receptor agonist, on the abuse liability of oxycodone among nondependent opioid users. Physiol Behav. 2016;159:33–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Ritz MC, Kuhar MJ. Relationship between self-administration of amphetamine and monoamine receptors in brain: comparison with cocaine. J Pharmacol Exp Ther. 1989;248(3):1010–7. [PubMed] [Google Scholar]
- 144.Sulzer D. How addictive drugs disrupt presynaptic dopamine neurotransmission. Neuron. 2011;69(4):628–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Ahmed SH, Lutjens R, van der Stap LD, Lekic D, Romano-Spica V, Morales M, et al. Gene expression evidence for remodeling of lateral hypothalamic circuitry in cocaine addiction. Proc Natl Acad Sci USA. 2005;102(32):11533–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Piechota M, Korostynski M, Solecki W, Gieryk A, Slezak M, Bilecki W, et al. The dissection of transcriptional modules regulated by various drugs of abuse in the mouse striatum. Genome Biol. 2010;11(5):R48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Lewitus GM, Konefal SC, Greenhalgh AD, Pribiag H, Augereau K, Stellwagen D. Microglial TNF-α suppresses cocaineinduced plasticity and behavioral sensitization. Neuron. 2016;90(3):483–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Costa BM, Yao H, Yang L, Buch S. Role of endoplasmic reticulum (ER) stress in cocaine-induced microglial cell death. J Neuroimmun Pharmacol. 2013;8(3):705–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Cao L, Walker MP, Vaidya NK, Fu M, Kumar S, Kumar A. Cocaine-mediated autophagy in astrocytes involves sigma 1 receptor, PI3K, mTOR, Atg5/7, beclin-1 and induces type II programmed cell death. Mol Neurobiol. 2016;53(7):4417–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Liao K, Guo M, Niu F, Yang L, Callen SE, Buch S. Cocainemediated induction of microglial activation involves the ER stress-TLR2 axis. J Neuroinflamm. 2016;13(1):33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Periyasamy P, Guo M-L, Buch S. Cocaine induces astrocytosis through ER stress-mediated activation of autophagy. Autophagy. 2016;12(8):1310–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Cotto B, Li H, Tuma RF, Ward SJ, Langford D. Cocaine-mediated activation of microglia and microglial MeCP2 and BDNF production. Neurobiol Dis. 2018;117:28–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Belin-Rauscent A, Lacoste J, Hermine O, Moussy A, Everitt BJ, Belin D. Decrease of cocaine, but not heroin, self-administration and relapse by the tyrosine kinase inhibitor masitinib in male Sprague Dawley rats. Psychopharmacology. 2018;235(5):1545–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.da Silva EZM, Jamur MC, Oliver C. Mast cell function: a new vision of an old cell. J Histochem Cytochem. 2014;62(10):698–738. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Northcutt AL, Hutchinson MR, Wang X, Baratta MV, Hiranita T, Cochran TA, et al. DAT isn’t all that: cocaine reward and reinforcement require Toll-like receptor 4 signaling. Mol Psychiatry. 2015;20(12):1525–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Gardner B, Zhu LX, Roth MD, Tashkin DP, Dubinett SM, Sharma S. Cocaine modulates cytokine and enhances tumor growth through sigma receptors. J Neuroimmunol. 2004;147(1):95–8. [DOI] [PubMed] [Google Scholar]
- 157.Lever JR, Fergason-Cantrell EA, Watkinson LD, Carmack TL, Lord SA, Xu R, et al. Cocaine occupancy of sigma1 receptors and dopamine transporters in mice. Synapse. 2016;70(3):98–111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Nguyen EC, McCracken KA, Liu Y, Pouw B, Matsumoto RR. Involvement of sigma (σ) receptors in the acute actions of methamphetamine: receptor binding and behavioral studies. Neuropharmacology. 2005;49(5):638–45. [DOI] [PubMed] [Google Scholar]
- 159.Sharkey J, Glen KA, Wolfe S, Kuhar MJ. Cocaine binding at σ receptors. Eur J Pharmacol. 1988;149(1):171–4. [DOI] [PubMed] [Google Scholar]
- 160.Yao H, Kim K, Duan M, Hayashi T, Guo M, Morgello S, et al. Cocaine hijacks σ1 receptor to initiate induction of activated leukocyte cell adhesion molecule: implication for increased monocyte adhesion and migration in the CNS. J Neurosci. 2011;31(16):5942–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Zhang Y, Lv X, Bai Y, Zhu X, Wu X, Chao J, et al. Involvement of sigma-1 receptor in astrocyte activation induced by methamphetamine via up-regulation of its own expression. J Neuroinflamm. 2015;12:29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Zhang Y, Zhu T, Zhang X, Chao J, Hu G, Yao H. Role of highmobility group box 1 in methamphetamine-induced activation and migration of astrocytes. J Neuroinflamm. 2015;12:156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Zhao J, Ha Y, Liou GI, Gonsalvez GB, Smith SB, Bollinger KE. Sigma receptor ligand, (+)-pentazocine, suppresses inflammatory responses of retinal microglia. Investig Ophthalmol Vis Sci. 2014;55(6):3375–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Shen K, Zhang Y, Lv X, Chen X, Zhou R, Nguyen LK, et al. Molecular mechanisms involving sigma-1 receptor in cell apoptosis of BV-2 microglial cells induced by methamphetamine. CNS Neurol Disord Drug Targets. 2016;15(7):857–65. [DOI] [PubMed] [Google Scholar]
- 165.Fernandes NC, Sriram U, Gofman L, Cenna JM, Ramirez SH, Potula R. Methamphetamine alters microglial immune function through P2X7R signaling. J Neuroinflamm. 2016;13(1):91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Wang B, Chen T, Wang J, Jia Y, Ren H, Wu F, et al. Methamphetamine modulates the production of interleukin-6 and tumor necrosis factor-alpha via the cAMP/PKA/CREB signaling pathway in lipopolysaccharide-activated microglia. Int Immunopharmacol. 2018;56:168–78. [DOI] [PubMed] [Google Scholar]
- 167.Mandyam CD, Koob GF. The addicted brain craves new neurons: putative role for adult-born progenitors in promoting recovery. Trends Neurosci. 2012;35(4):250–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Perry JL, Joseph JE, Jiang Y, Zimmerman RS, Kelly TH, Darna M, et al. Prefrontal cortex and drug abuse vulnerability: translation to prevention and treatment interventions. Brain Res Rev. 2011;65(2):124–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Anderson MA, Ao Y, Sofroniew MV. Heterogeneity of reactive astrocytes. Neurosci Lett. 2014;565:23–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Armstrong V, Reichel CM, Doti JF, Crawford CA, McDougall SA. Repeated amphetamine treatment causes a persistent elevation of glial fibrillary acidic protein in the caudate-putamen. Eur J Pharmacol. 2004;488(1):111–5. [DOI] [PubMed] [Google Scholar]
- 171.Bowers MS, Kalivas PW. Forebrain astroglial plasticity is induced following withdrawal from repeated cocaine administration. Eur J Neurosci. 2003;17(6):1273–8. [DOI] [PubMed] [Google Scholar]
- 172.Cearley CN, Blindheim K, Sorg BA, Krueger JM, Churchill L. Acute cocaine increases interleukin-1β mRNA and immunoreactive cells in the cortex and nucleus accumbens. Neurochem Res. 2011;36(4):686–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Gonçalves J, Martins T, Ferreira R, Milhazes N, Borges F, Ribeiro CF, et al. Methamphetamine-induced early increase of IL-6 and TNF-α mRNA expression in the mouse bain. Ann NY Acad Sci. 2008;1139(1):103–11. [DOI] [PubMed] [Google Scholar]
- 174.Berke JD, Hyman SE. Addiction, dopamine, and the molecular mechanisms of memory. Neuron. 2000;25(3):515–32. [DOI] [PubMed] [Google Scholar]
- 175.Pubill D, Canudas AM, Pallàs M, Camins A, Camarasa J, Escubedo E. Different glial response to methamphetamine- and methylenedioxymethamphetamine-induced neurotoxicity. Naunyn-Schmiedebergs Arch Pharmacol. 2003;367(5):490–9. [DOI] [PubMed] [Google Scholar]
- 176.Volkow ND, Wang G-J, Tomasi D, Baler RD. Unbalanced neuronal circuits in addiction. Curr Opin Neurobiol. 2013;23(4):639–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Krueger-Naug AMR, Hopkins DA, Armstrong JN, Plumier J-CL, Currie RW. Hyperthermic induction of the 27-kDa heat shock protein (Hsp27) in neuroglia and neurons of the rat central nervous system. J Comp Neurol. 2000;428(3):495–510. [DOI] [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(3):349–54. [DOI] [PubMed] [Google Scholar]
- 179.Epstein DH, Preston KL, Stewart J, Shaham Y. Toward a model of drug relapse: an assessment of the validity of the reinstatement procedure. Psychopharmacology. 2006;189(1):1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Hou Y-J, Yu ACH, Garcia JMRZ, Aotaki-Keen A, Lee Y-L, Eng LF, et al. Astrogliosis in culture. IV. Effects of basic fibroblast growth factor. J Neurosci Res. 1995;40(3):359–70. [DOI] [PubMed] [Google Scholar]
- 181.Flores C, Rodaros D, Stewart J. Long-lasting induction of astrocytic basic fibroblast growth factor by repeated injections of amphetamine: blockade by concurrent treatment with a glutamate antagonist. J Neurosci. 1998;18(22):9547–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Flores C, Samaha A-N, Stewart J. Requirement of endogenous basic fibroblast growth factor for sensitization to amphetamine. J Neurosci. 2000;20(2):RC55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Narita M, Miyatake M, Shibasaki M, Tsuda M, Koizumi S, Narita M, et al. Long-lasting change in brain dynamics induced by methamphetamine: enhancement of protein kinase C-dependent astrocytic response and behavioral sensitization. J Neurochem. 2005;93(6):1383–92. [DOI] [PubMed] [Google Scholar]
- 184.Thomas DM, Walker PD, Benjamins JA, Geddes TJ, Kuhn DM. Methamphetamine neurotoxicity in dopamine nerve endings of the striatumi is associated with microglial activation. J Pharmacol Exp Ther. 2004;311(1):1–7. [DOI] [PubMed] [Google Scholar]
- 185.Zhang L, Kitaichi K, Fujimoto Y, Nakayama H, Shimizu E, Iyo M, et al. Protective effects of minocycline on behavioral changes and neurotoxicity in mice after administration of methamphetamine. Prog Neuropsychopharmacol Biol Psychiatry. 2006;30(8):1381–93. [DOI] [PubMed] [Google Scholar]
- 186.Riddle EL, Fleckenstein AE, Hanson GR. Mechanisms of methamphetamine-induced dopaminergic neurotoxicity. AAPS J. 2006;8(2):E413–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Willuhn I, Burgeno LM, Groblewski PA, Phillips PEM. Excessive cocaine use results from decreased phasic dopamine signaling in the striatum. Nat Neurosci. 2014;17(5):704–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Sekine Y, Ouchi Y, Sugihara G, Takei N, Yoshikawa E, Nakamura K, et al. Methamphetamine causes microglial activation in the brains of human abusers. J Neurosci. 2008;28(22):5756–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Narendran R, Lopresti BJ, Mason NS, Deuitch L, Paris J, Himes ML, et al. 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(30):9945–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.McCann UD, Wong DF, Yokoi F, Villemagne V, Dannals RF, Ricaurte GA. Reduced striatal dopamine transporter density in abstinent methamphetamine and methcathinone users: evidence from positron emission tomography studies with [11C]WIN35,428. J Neurosci. 1998;18(20):8417–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Cadet JL, Bisagno V. Glial-neuronal ensembles: partners in drug addiction-associated synaptic plasticity. Front Pharmacol. 2014;5:204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Lu L, Wang X, Wu P, Xu C, Zhao M, Morales M, et al. Role of ventral tegmental area glial cell line-derived neurotrophic factor in incubation of cocaine craving. Biol Psychiatry. 2009;66(2):137–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Yan Y, Yamada K, Niwa M, Nagai T, Nitta A, Nabeshima T. Enduring vulnerability to reinstatement of methamphetamineseeking behavior in glial cell line-derived neurotrophic factor mutant mice. FASEB J. 2007;21(9):1994–2004. [DOI] [PubMed] [Google Scholar]
- 194.Yan Y, Miyamoto Y, Nitta A, Muramatsu S-I, Ozawa K, Yamada K, et al. Intrastriatal gene delivery of GDNF persistently attenuates methamphetamine self-administration and relapse in mice. Int J Neuropsychopharmacol. 2013;16(7):1559–67. [DOI] [PubMed] [Google Scholar]
- 195.Petrulli JR, Kalish B, Nabulsi NB, Huang Y, Hannestad J, Morris ED. Systemic inflammation enhances stimulant-induced striatal dopamine elevation. Transl Psychiatry. 2017;7(3):e1076. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Boury-Jamot B, Halfon O, Magistretti PJ, Boutrel B. Lactate release from astrocytes to neurons contributes to cocaine memory formation. BioEssays. 2016;38(12):1266–73. [DOI] [PubMed] [Google Scholar]
- 197.Castellano P, Nwagbo C, Martinez LR, Eugenin EA. Methamphetamine compromises gap junctional communication in astrocytes and neurons. J Neurochem. 2016;137(4):561–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Dackis CA, Gold MS. New concepts in cocaine addiction: the dopamine depletion hypothesis. Neurosci Biobehav Rev. 1985;9(3):469–77. [DOI] [PubMed] [Google Scholar]
- 199.Cadet JL, Krasnova IN, Jayanthi S, Lyles J. Neurotoxicity of substituted amphetamines: molecular and cellular mechanisms. Neurotoxicol Res. 2007;11(3–4):183–202. [DOI] [PubMed] [Google Scholar]
- 200.Kaushal N, Matsumoto RR. Role of sigma receptors in methamphetamine-induced neurotoxicity. Curr Neuropharmacol. 2011;9(1):54–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Pereira FC, Cunha-Oliveira T, Viana SD, Travassos AS, Nunes S, Silva C, et al. Disruption of striatal glutamatergic/GABAergic homeostasis following acute methamphetamine in mice. Neurotoxicol Teratol. 2012;34(5):522–9. [DOI] [PubMed] [Google Scholar]
- 202.Scofield MD, Heinsbroek JA, Gipson CD, Kupchik YM, Spencer S, Smith ACW, et al. The nucleus accumbens: mechanisms of addiction across drug classes reflect the importance of glutamate homeostasis. Pharmacol Rev. 2016;68(3):816–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Scofield MD. Exploring the role of astroglial glutamate release and association with synapses in neuronal function and behavior. Biol Psychiatry. 2018;84(11):778–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Rothstein JD, Dykes-Hoberg M, Pardo CA, Bristol LA, Jin L, Kuncl RW, et al. Knockout of glutamate transporters reveals a major role for astroglial transport in excitotoxicity and clearance of glutamate. Neuron. 1996;16(3):675–86. [DOI] [PubMed] [Google Scholar]
- 205.Fischer-Smith KD, Houston ACW, Rebec GV. Differential effects of cocaine access and withdrawal on glutamate type 1 transporter expression in rat nucleus accumbens core and shell. Neuroscience. 2012;210:333–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Parsegian A, See RE. Dysregulation of dopamine and glutamate release in the prefrontal cortex and nucleus accumbens following methamphetamine self-administration and during reinstatement in rats. Neuropsychopharmacology. 2014;39(4):811–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Alshehri FS, Althobaiti YS, Sari Y. Effects of administered ethanol and methamphetamine on glial glutamate transporters in rat striatum and hippocampus. J Mol Neurosci. 2017;61(3):343–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Scofield MD, Li H, Siemsen BM, Healey KL, Tran PK, Woronoff N, et al. Cocaine self-administration and extinction leads to reduced glial fibrillary acidic protein expression and morphometric features of astrocytes in the nucleus accumbens core. Biol Psychiatry. 2016;80(3):207–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Reissner KJ, Kalivas PW. Using glutamate homeostasis as a target for treating addictive disorders. Behav Pharmacol. 2010;21(5–6):514–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Abulseoud OA, Miller JD, Wu J, Choi DS, Holschneider DP. Ceftriaxone upregulates the glutamate transporter in medial prefrontal cortex and blocks reinstatement of methamphetamine seeking in a condition place preference paradigm. Brain Res. 2012;1456:14–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Bechard AR, Hamor PU, Schwendt M, Knackstedt LA. The effects of ceftriaxone on cue-primed reinstatement of cocaineseeking in male and female rats: estrous cycle effects on behavior and protein expression in the nucleus accumbens. Psychopharmacology. 2018;235(3):837–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Knackstedt LA, Melendez RI, Kalivas PW. Ceftriaxone restores glutamate homeostasis and prevents relapse to cocaine seeking. Biol Psychiatry. 2010;67(1):81–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.LaCrosse AL, Hill K, Knackstedt LA. Ceftriaxone attenuates cocaine relapse after abstinence through modulation of nucleus accumbens AMPA subunit expression. Eur Neuropsychopharmacol. 2016;26(2):186–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Niedzielska-Andres E, Mizera J, Sadakierska-Chudy A, Pomierny-Chamioło L, Filip M. Changes in the glutamate biomarker expression in rats vulnerable or resistant to the rewarding effects of cocaine and their reversal by ceftriaxone. Behav Brain Res. 2019;370:111945. [DOI] [PubMed] [Google Scholar]
- 215.Sari Y, Smith KD, Ali PK, Rebec GV. Upregulation of GLT1 attenuates cue-induced reinstatement of cocaine-seeking behavior in rats. J Neurosci. 2009;29(29):9239–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Madayag A, Lobner D, Kau KS, Mantsch JR, Abdulhameed O, Hearing M, et al. Repeated N-acetylcysteine administration alters plasticity-dependent effects of cocaine. J Neurosci. 2007;27(51):13968–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Moussawi K, Pacchioni A, Moran M, Olive MF, Gass JT, Lavin A, et al. N-Acetylcysteine reverses cocaine-induced metaplasticity. Nat Neurosci. 2009;12(2):182–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.Moussawi K, Zhou W, Shen H, Reichel CM, See RE, Carr DB, Kalivas PW. Reversing cocaine-induced synaptic potentiation provides enduring protection from relapse. Proc Natl Acad Sci USA. 2011;108(1):385–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Baker DA, McFarland K, Lake RW, Shen H, Tang X-C, Toda S, et al. Neuroadaptations in cystine–glutamate exchange underlie cocaine relapse. Nat Neurosci. 2003;6(7):743–9. [DOI] [PubMed] [Google Scholar]
- 220.Jastrzębska J, Frankowska M, Filip M, Atlas D. N-Acetylcysteine amide (AD4) reduces cocaine-induced reinstatement. Psychopharmacology. 2016;233(18):3437–48. [DOI] [PubMed] [Google Scholar]
- 221.Murray JE, Everitt BJ, Belin D. N-Acetylcysteine reduces early- and late-stage cocaine seeking without affecting cocaine taking in rats. Addict Biol. 2012;17(2):437–40. [DOI] [PubMed] [Google Scholar]
- 222.Reichel CM, Moussawi K, Do PH, Kalivas PW, See RE. Chronic N-acetylcysteine during abstinence or extinction after cocaine self-administration produces enduring reductions in drug seeking. J Pharmacol Exp Ther. 2011;337(2):487–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Siemsen BM, Reichel CM, Leong KC, Garcia-Keller C, Gipson CD, Spencer S, et al. Effects of methamphetamine self-administration and extinction on astrocyte structure and function in the nucleus accumbens core. Neuroscience. 2019;406:528–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224.Ward SJ, Rasmussen BA, Corley G, Henry C, Kim JK, Walker EA, et al. Beta-lactam antibiotic decreases acquisition of and motivation to respond for cocaine, but not sweet food, in C57Bl/6 mice. Behav Pharmacol. 2011;22(4):370–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225.Ducret E, Puaud M, Lacoste J, Belin-Rauscent A, Fouyssac M, Dugast E, et al. N-Acetylcysteine facilitates self-imposed abstinence after escalation of cocaine intake. Biol Psychiatry. 2016;80(3):226–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.Charntikov S, Pittenger ST, Pudiak CM, Bevins RA. The effect of N-acetylcysteine or bupropion on methamphetamine selfadministration and methamphetamine-triggered reinstatement of female rats. Neuropharmacology. 2018;135:487–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.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(5):577–81. [DOI] [PubMed] [Google Scholar]
- 228.Mizoguchi H, Yamada K, Nabeshima T. Neuropsychotoxicity of abused drugs: involvement of matrix metalloproteinase-2 and -9 and tissue inhibitor of matrix metalloproteinase-2 in methamphetamine-induced behavioral sensitization and reward in rodents. J Pharmacol Sci. 2008;106(1):9–14. [DOI] [PubMed] [Google Scholar]
- 229.Snider SE, Hendrick ES, Beardsley PM. Glial cell modulators attenuate methamphetamine self-administration in the rat. Eur J Pharmacol. 2013;701(1):124–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.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;4(53):142–8. 10.1016/j.pnpbp.2014.04.008 (Epub 2014 Apr 24). [DOI] [PubMed] [Google Scholar]
- 231.Chen H, Uz T, Manev H. Minocycline affects cocaine sensitization in mice. Neurosci Lett. 2009;452(3):258–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232.Poland RS, Hahn Y, Knapp PE, Beardsley PM, Bowers MS. Ibudilast attenuates expression of behavioral sensitization to cocaine in male and female rats. Neuropharmacology. 2016;109:281–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233.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(1):102–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234.Snider SE, Vunck SA, van den Oord EJCG, 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(1):75–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235.Reissner KJ, Brown RM, Spencer S, Tran PK, Thomas CA, Kalivas PW. Chronic administration of the methylxanthine propentofylline impairs reinstatement to cocaine by a GLT-1-dependent mechanism. Neuropsychopharmacology. 2014;39(2):499–506. 10.1038/npp.2013.223 (Epub 2013 Aug 29). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236.Miller WR, Fox RG, Stutz SJ, Lane SD, Denner L, Cunningham KA, et al. PPARγ agonism attenuates cocaine cue reactivity. Addict Biol. 2018;23(1):55–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237.Thomsen M, Caine SB. Psychomotor stimulant effects of cocaine in rats and 15 mouse strains. Exp Clin Psychopharmacol. 2011;19(5):321–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238.Ciraulo DA, Sarid-Segal O, Knapp CM, Ciraulo AM, LoCastro J, Bloch DA, et al. Efficacy screening trials of paroxetine, pentoxifylline, riluzole, pramipexole and venlafaxine in cocaine dependence. Addiction. 2005;100(Suppl. 1):12–22. [DOI] [PubMed] [Google Scholar]
- 239.Reid MS, Angrist B, Baker S, Woo C, Schwartz M, Montgomery A, et al. A placebo-controlled screening trial of celecoxib for the treatment of cocaine dependence. Addiction. 2005;100(s1):32–42. [DOI] [PubMed] [Google Scholar]
- 240.Santos-Galdiano M, Pérez-Rodríguez D, Anuncibay-Soto B, Font-Belmonte E, Ugidos IF, Pérez-García CC, et al. Celecoxib treatment improves neurologic deficit and reduces selective neuronal loss and glial response in rats after transient middle cerebral artery occlusion. J Pharmacol Exp Ther. 2018;367(3):528–42. [DOI] [PubMed] [Google Scholar]
- 241.Schmitz JM, Green CE, Hasan KM, Vincent J, Suchting R, Weaver MF, et al. PPAR-gamma agonist pioglitazone modifies craving intensity and brain white matter integrity in patients with primary cocaine use disorder: a double-blind randomized controlled pilot trial. Addiction. 2017;112(10):1861–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242.Xu J, DeVito EE, Worhunsky PD, Carroll KM, Rounsaville BJ, Potenza MN. White matter integrity is associated with treatment outcome measures in cocaine dependence. Neuropsychopharmacology. 2010;35(7):1541–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 243.Moeller FG, Hasan KM, Steinberg JL, Kramer LA, Dougherty DM, Santos RM, et al. Reduced anterior corpus callosum white matter integrity is related to increased impulsivity and reduced discriminability in cocaine-dependent subjects: diffusion tensor imaging. Neuropsychopharmacology. 2005;30(3):610–7. [DOI] [PubMed] [Google Scholar]
- 244.Mardikian PN, LaRowe SD, Hedden S, Kalivas PW, Malcolm RJ. An open-label trial of N-acetylcysteine for the treatment of cocaine dependence: a pilot study. Prog Neuropsychopharmacol Biol Psychiatry. 2007;31(2):389–94. [DOI] [PubMed] [Google Scholar]
- 245.LaRowe SD, Myrick H, Hedden S, Mardikian P, Saladin M, McRae A, et al. Is cocaine desire reduced by N-acetylcysteine? Am J Psychiatry. 2007;164(7):1115–7. [DOI] [PubMed] [Google Scholar]
- 246.Amen SL, Piacentine LB, Ahmad ME, Li S-J, Mantsch JR, Risinger RC, et al. Repeated N-acetyl cysteine reduces cocaine seeking in rodents and craving in cocaine-dependent humans. Neuropsychopharmacology. 2010;36:871. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247.LaRowe SD, Kalivas PW, Nicholas JS, Randall PK, Mardikian PN, Malcolm RJ. A double-blind placebo-controlled trial of N-acetylcysteine in the treatment of cocaine dependence. Am J Addict. 2013;22(5):443–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248.Back SE, McCauley JL, Korte KJ, Gros DF, Leavitt V, Gray KM, et al. A double-blind, randomized, controlled pilot trial of N-acetylcysteine in veterans with posttraumatic stress disorder and substance use disorders. J Clin Psychiatry. 2016;77(11):e1439–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 249.Levi Bolin B, Alcorn JL 3rd, Lile JA, Rush CR, Rayapati AO, Hays LR, et al. N-Acetylcysteine reduces cocaine-cue attentional bias and differentially alters cocaine self-administration based on dosing order. Drug Alcohol Depend. 2017;1(178):452–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250.Duailibi MS, Cordeiro Q, Brietzke E, Ribeiro M, LaRowe S, Berk M, et al. N-Acetylcysteine in the treatment of craving in substance use disorders: systematic review and meta-analysis. Am J Addict. 2017;26(7):660–6. [DOI] [PubMed] [Google Scholar]
- 251.Echevarria MA Nocito, Andrade Reis T, Ruffo Capatti G, Siciliano Soares V, da Silveira DX, Fidalgo TM. N-Acetylcysteine for treating cocaine addiction: a systematic review. Psychiatry Res. 2017;251:197–203. [DOI] [PubMed] [Google Scholar]
- 252.Mousavi SG, Sharbafchi MR, Salehi M, Peykanpour M, Karimian Sichani N, Maracy M. The efficacy of N-acetylcysteine in the treatment of methamphetamine dependence: a double-blind controlled, crossover study. Arch Iran Med. 2015;18(1):28–33. [PubMed] [Google Scholar]
- 253.Worley MJ, Heinzerling KG, Roche DJO, Shoptaw S. Ibudilast attenuates subjective effects of methamphetamine in a placebo-controlled inpatient study. Drug Alcohol Depend. 2016;162:245–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 254.Birath JB, Briones M, Amaya S, Shoptaw S, Swanson A-N, Tsuang J, et al. Ibudilast may improve attention during early abstinence from methamphetamine. Drug Alcohol Depend. 2017;178:386–90. [DOI] [PubMed] [Google Scholar]
- 255.Sofuoglu M, Mooney M, Kosten T, Waters A, Hashimoto K. Minocycline attenuates subjective rewarding effects of dextroamphetamine in humans. Psychopharmacology. 2011;213(1):61–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 256.Vengeliene V, Bilbao A, Molander A, Spanagel R. Neuropharmacology of alcohol addiction. Br J Pharmacol. 2008;154(2):299–315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257.Blanco AM, Vallés SL, Pascual M, Guerri C. Involvement of TLR4/type I IL-1 receptor signaling in the induction of inflammatory mediators and cell death induced by ethanol in cultured astrocytes. J Immunol. 2005;175(10):6893–9. [DOI] [PubMed] [Google Scholar]
- 258.Davis RL, Syapin PJ. Ethanol increases nuclear factorkappa B activity in human astroglial cells. Neurosci Lett. 2004;371(2–3):128–32. [DOI] [PubMed] [Google Scholar]
- 259.Zou J, Crews F. CREB and NF-κB Transcription factors regulate sensitivity to excitotoxic and oxidative stress induced neuronal cell death. Cell Mol Neurobiol. 2006;26(4):383–403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 260.Alfonso-Loeches S, Pascual-Lucas M, Blanco AM, SanchezVera I, Guerri C. Pivotal role of TLR4 receptors in alcoholinduced neuroinflammation and brain damage. J Neurosci. 2010;30(24):8285–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 261.Boyadjieva NI, Sarkar DK. Role of microglia in ethanol’s apoptotic action on hypothalamic neuronal cells in primary cultures. Alcohol Clin Exp Res. 2010;34(11):1835–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 262.Guerri C, Sáez R, Sancho-Tello M, Martin de Aquilera E, Renau-Piqueras J. Ethanol alters astrocyte development: a study of critical periods using primary cultures. Neurochem Res. 1990;15(5):559–65. [DOI] [PubMed] [Google Scholar]
- 263.Vetreno RP, Crews FT. Adolescent binge drinking increases expression of the danger signal receptor agonist HMGB1 and Toll-like receptors in the adult prefrontal cortex. Neuroscience. 2012;226:475–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 264.Bajo M, Varodayan FP, Madamba SG, Robert AJ, Casal LM, Oleata CS, et al. IL-1 interacts with ethanol effects on GABAergic transmission in the mouse central amygdala. Front Pharmacol. 2015;19(6):49. 10.3389/fphar.2015.00049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 265.Franke H. Influence of chronic alcohol treatment on the GFAPimmunoreactivity in astrocytes of the hippocampus in rats. Acta Histochem. 1995;97(3):263–71. [DOI] [PubMed] [Google Scholar]
- 266.Marshall SA, McClain JA, Kelso ML, Hopkins DM, Pauly JR, Nixon K. Microglial activation is not equivalent to neuroinflammation in alcohol-induced neurodegeneration: the importance of microglia phenotype. Neurobiol Dis. 2013;54:239–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 267.Dlugos CA, Pentney RJ. Quantitative immunocytochemistry of glia in the cerebellar cortex of old ethanol-fed rats. Alcohol. 2001;23(2):63–9. [DOI] [PubMed] [Google Scholar]
- 268.Kane CJM, Phelan KD, Douglas JC, Wagoner G, Johnson JW, Xu J, et al. Effects of ethanol on immune response in the brain: region-specific changes in aged mice. J Neuroinflamm. 2013;10(1):834. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 269.Kane CJM, Phelan KD, Douglas JC, Wagoner G, Johnson JW, Xu J, et al. Effects of ethanol on immune response in the brain: region-specific changes in adolescent versus adult mice. Alcohol Clin Exp Res. 2014;38(2):384–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 270.Qin L, He J, Hanes RN, Pluzarev O, Hong J-S, Crews FT. Increased systemic and brain cytokine production and neuroinflammation by endotoxin following ethanol treatment. J Neuroinflamm. 2008;5(1):10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 271.Zahr NM, Luong R, Sullivan EV, Pfefferbaum A. Measurement of serum, liver, and brain cytokine induction, thiamine levels, and hepatopathology in rats exposed to a 4-day alcohol binge protocol. Alcohol Clin Exp Res. 2010;34(11):1858–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 272.Korbo L. Glial cell loss in the hippocampus of alcoholics. Alcohol Clin Exp Res. 1999;23(1):164–8. [PubMed] [Google Scholar]
- 273.Miguel-Hidalgo JJ, Wei J, Andrew M, Overholser JC, Jurjus G, Stockmeier CA, et al. Glia pathology in the prefrontal cortex in alcohol dependence with and without depressive symptoms. Biol Psychiatry. 2002;52(12):1121–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 274.He J, Crews FT. Increased MCP-1 and microglia in various regions of the human alcoholic brain. Exp Neurol. 2008;210(2):349–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 275.Liu J, Lewohl JM, Harris RA, Iyer VR, Dodd PR, Randall PK, et al. Patterns of gene expression in the frontal cortex discriminate alcoholic from nonalcoholic individuals. Neuropsychopharmacology. 2006;31(7):1574–82. [DOI] [PubMed] [Google Scholar]
- 276.Yakovleva T, Bazov I, Watanabe H, Hauser KF, Bakalkin G. Transcriptional control of maladaptive and protective responses in alcoholics: a role of the NF-κB system. Brain Behav Immun. 2011;25(Suppl. 1):S29–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 277.Fernandez-Lizarbe S, Montesinos J, Guerri C. Ethanol induces TLR4/TLR2 association, triggering an inflammatory response in microglial cells. J Neurochem. 2013;126(2):261–73. [DOI] [PubMed] [Google Scholar]
- 278.Pascual M, Baliño P, Alfonso-Loeches S, Aragón CM, Guerri C. Impact of TLR4 on behavioral and cognitive dysfunctions associated with alcohol-induced neuroinflammatory damage. Brain Behav Immun. 2011;25(Suppl. 1):S80–91. [DOI] [PubMed] [Google Scholar]
- 279.Alfonso-Loeches S, Pascual M, Guerri C. Gender differences in alcohol-induced neurotoxicity and brain damage. Toxicology. 2013;311(1–2):27–34. [DOI] [PubMed] [Google Scholar]
- 280.Corrigan F, Wu Y, Tuke J, Coller JK, Rice KC, Diener KR, et al. Alcohol-induced sedation and synergistic interactions between alcohol and morphine: a key mechanistic role for Toll-like receptors and MyD88-dependent signaling. Brain Behav Immun. 2015;45:245–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 281.Fernandez-Lizarbe S, Pascual M, Guerri C. Critical role of TLR4 response in the activation of microglia induced by ethanol. J Immunol. 2009;183(7):4733–44. [DOI] [PubMed] [Google Scholar]
- 282.Fernandez-Lizarbe S, Pascual M, Gascon MS, Blanco A, Guerri C. Lipid rafts regulate ethanol-induced activation of TLR4 signaling in murine macrophages. Mol Immunol. 2008;45(7):2007–16. [DOI] [PubMed] [Google Scholar]
- 283.Pascual M, Baliño P, Aragón CM, Guerri C. Cytokines and chemokines as biomarkers of ethanol-induced neuroinflammation and anxiety-related behavior: role of TLR4 and TLR2. Neuropharmacology. 2015;89:352–9. [DOI] [PubMed] [Google Scholar]
- 284.Vallés SL, Blanco AM, Pascual M, Guerri C. Chronic ethanol treatment enhances inflammatory mediators and cell death in the brain and in astrocytes. Brain Pathol. 2004;14(4):365–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 285.June HL, Liu J, Warnock KT, Bell KA, Balan I, Bollino D, et al. CRF-amplified neuronal TLR4/MCP-1 signaling regulates alcohol self-administration. Neuropsychopharmacology. 2015;40(6):1549–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 286.Liu J, Yang AR, Kelly T, Puche A, Esoga C, June HL Jr, et al. Binge alcohol drinking is associated with GABAA alpha2regulated Toll-like receptor 4 (TLR4) expression in the central amygdala. Proc Natl Acad Sci USA. 2011;108(11):4465–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 287.Blednov YA, Bergeson SE, Walker D, Ferreira VMM, Kuziel WA, Harris RA. Perturbation of chemokine networks by gene deletion alters the reinforcing actions of ethanol. Behav Brain Res. 2005;165(1):110–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 288.Al-Ofi EA, Al-Ghamdi BS. High-mobility group box 1 (HMGB1), an endogenous ligand of Toll-like receptors 2 and 4, induces astroglial inflammation via NF-beta pathway. Folia Morphol (Warsz). 2018;78(1):10–6. [DOI] [PubMed] [Google Scholar]
- 289.Lowe PP, Gyongyosi B, Satishchandran A, Iracheta-Vellve A, Cho Y, Ambade A, et al. Reduced gut microbiome protects from alcohol-induced neuroinflammation and alters intestinal and brain inflammasome expression. J Neuroinflamm. 2018;15(1):298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 290.Crews FT, Qin L, Sheedy D, Vetreno RP, Zou J. High mobility group box 1/Toll-like receptor danger signaling increases brain neuroimmune activation in alcohol dependence. Biol Psychiatry. 2013;73(7):602–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 291.Zou JY, Crews FT. Release of neuronal HMGB1 by ethanol through decreased HDAC activity activates brain neuroimmune signaling. PloS one. 2014;9:e87915. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 292.Perry VH, Holmes C. Microglial priming in neurodegenerative disease. Nat Rev Neurol. 2014;10:217. [DOI] [PubMed] [Google Scholar]
- 293.Drew PD, Kane CJM. Neuroimmune mechanisms of glia and their interplay with alcohol exposure across the lifespan. In: Cui C, Grandison L, Noronha A, editors. Neural-immune interactions in brain function and alcohol related disorders. Boston: Springer; 2013. p. 359–86. [Google Scholar]
- 294.Bhattacharya A, Derecki NC, Lovenberg TW, Drevets WC. Role of neuro-immunological factors in the pathophysiology of mood disorders. Psychopharmacology. 2016;233(9):1623–36. [DOI] [PubMed] [Google Scholar]
- 295.Muller N. The role of anti-inflammatory treatment in psychiatric disorders. Psychiatr Danub. 2013;25(3):292–8. [PubMed] [Google Scholar]
- 296.Heberlein A, Käser M, Lichtinghagen R, Rhein M, Lenz B, Kornhuber J, et al. TNF-α and IL-6 serum levels: neurobiological markers of alcohol consumption in alcohol-dependent patients? Alcohol. 2014;48(7):671–6. [DOI] [PubMed] [Google Scholar]
- 297.Leclercq S, De Saeger C, Delzenne N, de Timary P, Stärkel P. Role of inflammatory pathways, blood mononuclear cells, and gut-derived bacterial products in alcohol dependence. Biol Psychiatry. 2014;76(9):725–33. [DOI] [PubMed] [Google Scholar]
- 298.Breese GR, Knapp DJ, Overstreet DH, Navarro M, Wills TA, Angel RA. Repeated lipopolysaccharide (LPS) or cytokine treatments sensitize ethanol withdrawal-induced anxiety-like behavior. Neuropsychopharmacology. 2008;33(4):867–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 299.Freeman K, Brureau A, Vadigepalli R, Staehle MM, Brureau MM, Gonye GE, et al. Temporal changes in innate immune signals in a rat model of alcohol withdrawal in emotional and cardiorespiratory homeostatic nuclei. J Neuroinflamm. 2012;9(1):97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 300.Stevenson JR, Schroeder JP, Nixon K, Besheer J, Crews FT, Hodge CW. Abstinence following alcohol drinking produces depression-like behavior and reduced hippocampal neurogenesis in mice. Neuropsychopharmacology. 2009;34(5):1209–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 301.Agrawal RG, Hewetson A, George CM, Syapin PJ, Bergeson SE. Minocycline reduces ethanol drinking. Brain Behav Immun. 2011;25(Suppl. 1):S165–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 302.Gajbhiye SV, Tripathi RK, Salve B, Petare A, Potey AV. Evaluation of effect of minocycline on rewarding potential and alcohol relapse in place preference model in mice. Neurosci Lett. 2017;649:28–33. [DOI] [PubMed] [Google Scholar]
- 303.Harper KM, Knapp DJ, Park MA, Breese GR. Differential effects of single versus repeated minocycline administration: lack of significant interaction with chronic alcohol history. Pharmacol Biochem Behav. 2018;168:33–42. [DOI] [PubMed] [Google Scholar]
- 304.Plane JM, Shen Y, Pleasure DE, Deng W. Prospects for minocycline neuroprotection. Arch Neurol. 2010;67(12):1442–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 305.Bergeson SE, Nipper MA, Jensen J, Helms ML, Finn DA. Tigecycline reduces ethanol intake in dependent and nondependent male and female C57BL/6J mice. Alcohol Clin Exp Res. 2016;40(12):2491–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 306.Chen H, Wu J, Zhang J, Fujita Y, Ishima T, Iyo M, et al. Protective effects of the antioxidant sulforaphane on behavioral changes and neurotoxicity in mice after the administration of methamphetamine. Psychopharmacology. 2012;222(1):37–45. [DOI] [PubMed] [Google Scholar]
- 307.Cozzoli DK, Kaufman MN, Nipper MA, Hashimoto JG, Wiren KM, Finn DA. Functional regulation of PI3K-associated signaling in the accumbens by binge alcohol drinking in male but not female mice. Neuropharmacology. 2016;105:164–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 308.George FR. The role of arachidonic acid metabolites in mediating ethanol self-administration and intoxication. Ann N Y Acad Sci. 1989;559(1):382–91. [DOI] [PubMed] [Google Scholar]
- 309.Martinez JM, Groot JA, Curtis DC, Allison CL, Marquardt PC, Holmes AN, et al. Effective reduction of acute ethanol withdrawal by the tetracycline derivative, tigecycline, in female and male DBA/2J mice. Alcohol Clin Exp Res. 2016;40(12):2499–505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 310.Oliveros A, Choi D-S. Repurposing tigecycline for the treatment of alcohol use disorder. Alcohol Clin Exp Res. 2017;41(3):497–500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 311.Pla A, Pascual M, Guerri C. Autophagy constitutes a protective mechanism against ethanol toxicity in mouse astrocytes and neurons. PLoS One. 2016;11(4):e0153097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 312.Pascual M, Blanco AM, Cauli O, Miñarro J, Guerri C. Intermittent ethanol exposure induces inflammatory brain damage and causes long-term behavioural alterations in adolescent rats. Eur J Neurosci. 2007;25(2):541–50. [DOI] [PubMed] [Google Scholar]
- 313.Zou JY, Crews FT. Release of neuronal HMGB1 by ethanol through decreased HDAC activity activates brain neuroimmune signaling. PLoS One. 2014;9(2):e87915. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 314.Syapin PJ, Martinez JM, Curtis DC, Marquardt PC, Allison CL, Groot JA, et al. Effective reduction in high ethanol drinking by semisynthetic tetracycline derivatives. Alcohol Clin Exp Res. 2016;40(12):2482–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 315.Lee MR, Ruby CL, Hinton DJ, Choi S, Adams CA, Young Kang N, et al. Striatal adenosine signaling regulates EAAT2 and astrocytic AQP4 expression and alcohol drinking in mice. Neuropsychopharmacology. 2013;38(3):437–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 316.Qrunfleh AM, Alazizi A, Sari Y. Ceftriaxone, a beta-lactam antibiotic, attenuates relapse-like ethanol-drinking behavior in alcohol-preferring rats. J Psychopharmacol. 2013;27(6):541–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 317.Rao P, Sari Y. Effectiveness of ceftriaxone treatment in preventing relapse-like drinking behavior following long-term ethanol dependence in P rats. J Addict Res Ther. 2014;5:1000183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 318.Weiland A, Garcia S, Knackstedt LA. Ceftriaxone and cefazolin attenuate the cue-primed reinstatement of alcohol-seeking. Front Pharmacol. 2015;6:44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 319.Bell RL, Lopez MF, Cui C, Egli M, Johnson KW, Franklin KM, et al. Ibudilast reduces alcohol drinking in multiple animal models of alcohol dependence. Addict Biol. 2015;20(1):38–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 320.Blednov YA, Benavidez JM, Black M, Harris RA. Inhibition of phosphodiesterase 4 reduces ethanol intake and preference in C57BL/6J mice. Front Neurosci. 2014;8:129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 321.Hu W, Lu T, Chen A, Huang Y, Hansen R, Chandler LJ, et al. Inhibition of phosphodiesterase-4 decreases ethanol intake in mice. Psychopharmacology. 2011;218(2):331–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 322.Wen R-T, Zhang M, Qin W-J, Liu Q, Wang W-P, Lawrence AJ, et al. The phosphodiesterase-4 (PDE4) inhibitor rolipram decreases ethanol seeking and consumption in alcohol-preferring Fawn-Hooded rats. Alcohol Clin Exp Res. 2012;36(12):2157–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 323.Drew PD, Johnson JW, Douglas JC, Phelan KD, Kane CJM. Pioglitazone blocks ethanol induction of microglial activation and immune responses in the hippocampus, cerebellum, and cerebral cortex in a mouse model of fetal alcohol spectrum disorders. Alcohol Clin Exp Res. 2015;39(3):445–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 324.Kane CJM, Phelan KD, Han L, Smith RR, Xie J, Douglas JC, et al. Protection of neurons and microglia against ethanol in a mouse model of fetal alcohol spectrum disorders by peroxisome proliferator-activated receptor-γ agonists. Brain Behav Immun. 2011;25:S137–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 325.Cippitelli A, Domi E, Ubaldi M, Douglas JC, Li HW, Demopulos G, et al. Protection against alcohol-induced neuronal and cognitive damage by the PPARγ receptor agonist pioglitazone. Brain Behav Immun. 2017;64:320–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 326.Blednov YA, Benavidez JM, Black M, Ferguson LB, Schoenhard GL, Goate AM, et al. Peroxisome proliferator-activated receptors α and γ are linked with alcohol consumption in mice and withdrawal and dependence in humans. Alcohol Clin Exp Res. 2015;39(1):136–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 327.Ferguson LB, Most D, Blednov YA, Harris RA. PPAR agonists regulate brain gene expression: relationship to their effects on ethanol consumption. Neuropharmacology. 2014;86:397–407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 328.Stopponi S, Somaini L, Cippitelli A, Cannella N, Braconi S, Kallupi M, et al. Activation of nuclear PPARγ receptors by the antidiabetic agent pioglitazone suppresses alcohol drinking and relapse to alcohol seeking. Biol Psychiatry. 2011;69(7):642–9. [DOI] [PubMed] [Google Scholar]
- 329.Stopponi S, de Guglielmo G, Somaini L, Cippitelli A, Cannella N, Kallupi M, et al. Activation of PPARγ by pioglitazone potentiates the effects of naltrexone on alcohol drinking and relapse in msP rats. Alcohol Clin Exp Res. 2013;37(8):1351–60. [DOI] [PubMed] [Google Scholar]
- 330.Ferreira Seiva FR, Amauchi JF, Ribeiro Rocha KK, Souza GA, Ebaid GX, Burneiko RM, et al. Effects of N-acetylcysteine on alcohol abstinence and alcohol-induced adverse effects in rats. Alcohol. 2009;43(2):127–35. [DOI] [PubMed] [Google Scholar]
- 331.Mocelin R, Marcon M, da Rosa Araujo AS, Herrmann AP, Piato A. Withdrawal effects following repeated ethanol exposure are prevented by N-acetylcysteine in zebrafish. Prog Neuropsychopharmacol Biol Psychiatry. 2019;93:161–70. [DOI] [PubMed] [Google Scholar]
- 332.Lebourgeois S, González-Marín MC, Antol J, Naassila M, Vilpoux C. Evaluation of N-acetylcysteine on ethanol selfadministration in ethanol-dependent rats. Neuropharmacology. 2019;150:112–20. [DOI] [PubMed] [Google Scholar]
- 333.Ray LA, Bujarski S, Shoptaw S, Roche DJ, Heinzerling K, Miotto K. Development of the neuroimmune modulator ibudilast for the treatment of alcoholism: a randomized, placebocontrolled, human laboratory trial. Neuropsychopharmacology. 2017;42(9):1776–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 334.Petrakis IL, Ralevski E, Gueorguieva R, Sloan ME, Devine L, Yoon G, et al. Targeting neuroinflammation with minocycline in heavy drinkers. Psychopharmacology (Berl). 2019;236(10):3013–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 335.Kohno M, Link J, Dennis LE, McCready H, Huckans M, Hoffman WF, et al. Neuroinflammation in addiction: a review of neuroimaging studies and potential immunotherapies. Pharmacol Biochem Behav. 2019;179:34–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 336.Sopori M. Effects of cigarette smoke on the immune system. Nat Rev Immunol. 2002;2(5):372–7. [DOI] [PubMed] [Google Scholar]
- 337.Geng Y, Savage SM, Razani-Boroujerdi S, Sopori ML. Effects of nicotine on the immune response. II. Chronic nicotine treatment induces T cell anergy. J Immunol. 1996;156(7):2384–90. [PubMed] [Google Scholar]
- 338.Egea J, Buendia I, Parada E, Navarro E, León R, Lopez MG. Anti-inflammatory role of microglial alpha7 nAChRs and its role in neuroprotection. Biochem Pharmacol. 2015;97(4):463–72. [DOI] [PubMed] [Google Scholar]
- 339.Guan Y-Z, Jin X-D, Guan L-X, Yan H-C, Wang P, Gong Z, et al. Nicotine inhibits microglial proliferation and is neuroprotective in global ischemia rats. Mol Neurobiol. 2015;51(3):1480–8. [DOI] [PubMed] [Google Scholar]
- 340.Noda M, Kobayashi AI. Nicotine inhibits activation of microglial proton currents via interactions with α7 acetylcholine receptors. J Physiol Sci. 2017;67(1):235–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 341.Wang H, Yu M, Ochani M, Amella CA, Tanovic M, Susarla S, et al. Nicotinic acetylcholine receptor α7 subunit is an essential regulator of inflammation. Nature. 2003;421(6921):384–8. [DOI] [PubMed] [Google Scholar]
- 342.Westerlund A, Björklund U, Rönnbäck L, Hansson E. Long-term nicotine treatment suppresses IL-1β release and attenuates substance P- and 5-HT-evoked Ca2 + responses in astrocytes. Neurosci Lett. 2013;553:191–5. [DOI] [PubMed] [Google Scholar]
- 343.Brody AL, Hubert R, Enoki R, Garcia LY, Mamoun MS, Okita K, et al. Effect of cigarette smoking on a marker for neuroinflammation: a [(11)C]DAA1106 positron emission tomography study. Neuropsychopharmacology. 2017;42(8):1630–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 344.Fields RD. Myelin: more than insulation. Science. 2014;344(6181):264–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 345.Paul RH, Grieve SM, Niaura R, David SP, Laidlaw DH, Cohen R, et al. Chronic cigarette smoking and the microstructural integrity of white matter in healthy adults: a diffusion tensor imaging study. Nicotine Tob Res. 2008;10(1):137–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 346.López-Hidalgo M, Salgado-Puga K, Alvarado-Martínez R, Medina AC, Prado-Alcalá RA, García-Colunga J. Nicotine uses neuron-glia communication to enhance hippocampal synaptic transmission and long-term memory. PLoS One. 2012;7(11):e49998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 347.Cao J, Dwyer JB, Gautier NM, Leslie FM, Li MD. Central myelin gene expression during postnatal development in rats exposed to nicotine gestationally. Neurosci Lett. 2013;553:115–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 348.Martínez-Rodríguez R, Toledano A, Álvarez MI, Turégano L, Colman O, Rosés P, et al. Chronic nicotine administration increases NGF-like immunoreactivity in frontoparietal cerebral cortex. J Neurosci Res. 2003;73(5):708–16. [DOI] [PubMed] [Google Scholar]
- 349.Park HJ, Lee PH, Ahn YW, Choi YJ, Lee G, Lee D-Y, et al. Neuroprotective effect of nicotine on dopaminergic neurons by anti-inflammatory action. Eur J Neurosci. 2007;26(1):79–89. [DOI] [PubMed] [Google Scholar]
- 350.Quik M, Campos C, Parameswaran N, Langston JW, McIntosh JM, Yeluashvili M. Chronic nicotine treatment increases nAChRs and microglial expression in monkey substantia nigra after nigrostriatal damage. J Mol Neurosci. 2010;40(1–2):105–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 351.Alajaji M, Bowers MS, Knackstedt L, Damaj MI. Effects of the beta-lactam antibiotic ceftriaxone on nicotine withdrawal and nicotine-induced reinstatement of preference in mice. Psychopharmacology. 2013;228(3):419–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 352.Philogene-Khalid HL, Simmons SJ, Muschamp JW, Rawls SM. Effects of ceftriaxone on conditioned nicotine reward in rats. Behav Pharmacol. 2017;28(6):485–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 353.Sari Y, Toalston JE, Rao PSS, Bell RL. Effects of ceftriaxone on ethanol, nicotine or sucrose intake by alcohol-preferring (P) rats and its association with GLT-1 expression. Neuroscience. 2016;326:117–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 354.Bowers MS, Jackson A, Maldoon PP, Damaj MI. N-Acetylcysteine decreased nicotine reward-like properties and withdrawal in mice. Psychopharmacology. 2016;233(6):995–1003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 355.Ramirez-Niño AM, D’Souza MS, Markou A. N-Acetylcysteine decreased nicotine self-administration and cue-induced reinstatement of nicotine seeking in rats: comparison with the effects of N-acetylcysteine on food responding and food seeking. Psychopharmacology. 2013;225(2):473–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 356.Namba MD, Kupchik YM, Spencer SM, Garcia-Keller C, Goenaga JG, Powell GL, et al. Accumbens neuroimmune signaling and dysregulation of astrocytic glutamate transport underlie conditioned nicotine-seeking behavior. Addict Biol. e12797. (Epub 2019 Jul 22). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 357.Goenaga J, Powell GL, Leyrer-Jackson JM, Piña J, Phan S, Prakapenka AV, et al. N-Acetylcysteine yields sex-specific efficacy for cue-induced reinstatement of nicotine seeking. Addict Biol. 2020;25(1):e12711. 10.1111/adb.12711 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 358.Moro F, Giannotti G, Caffino L, Marzo CM, Di Clemente A, Fumagalli F, et al. Lasting reduction of nicotine-seeking behavior by chronic N-acetylcysteine during experimental cue-exposure therapy. Addict Biol. 2019;27:e12771. [DOI] [PubMed] [Google Scholar]
- 359.Powell GL, Leyrer-Jackson JM, Goenaga J, Namba MD, Piña J, Spencer S, et al. Chronic treatment with N-acetylcysteine decreases extinction responding and reduces cue-induced nicotine-seeking. Physiol Rep. 2019;7(1):e13958. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 360.McClure EA, Baker NL, Gipson CD, Carpenter MJ, Roper AP, Froeliger BE, et al. An open-label pilot trial of N-acetylcysteine and varenicline in adult cigarette smokers. Am J Drug Alcohol Abuse. 2015;41(1):52–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 361.Froeliger B, McConnell PA, Stankeviciute N, McClure EA, Kalivas PW, Gray KM. The effects of N-acetylcysteine on frontostriatal resting-state functional connectivity, withdrawal symptoms and smoking abstinence: a double-blind, placebo-controlled fMRI pilot study. Drug Alcohol Depend. 2015;156:234–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 362.Grant JE, Odlaug BL, Chamberlain SR, Potenza MN, Schreiber LR, Donahue CB, et al. A randomized, placebo-controlled trial of N-acetylcysteine plus imaginal desensitization for nicotine-dependent pathological gamblers. J Clin Psychiatry. 2014;75(1):39–45. [DOI] [PubMed] [Google Scholar]
- 363.Knackstedt LA, LaRowe S, Mardikian P, Malcolm R, Upadhyaya H, Hedden S, Markou A, Kalivas PW. The role of cystine-glutamate exchange in nicotine dependence in rats and humans. Biol Psychiatry. 2009;65(10):841–5. 10.1016/j.biopsych.2008.10.040 (Epub 2008 Dec 21). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 364.Jones JD, Comer SD, Metz VE, Manubay JM, Mogali S, Ciccocioppo R, et al. Pioglitazone, a PPARγ agonist, reduces nicotine craving in humans, with marginal effects on abuse potential. Pharmacol Biochem Behav. 2017;163:90–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 365.Sofuoglu M, Waters AJ, Mooney M, O’Malley SS. Minocycline reduced craving for cigarettes but did not affect smoking or intravenous nicotine responses in humans. Pharmacol Biochem Behav. 2009;92(1):135–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 366.Jean-Gilles L, Gran B, Constantinescu CS. Interaction between cytokines, cannabinoids and the nervous system. Immunobiology. 2010;215(8):606–10. [DOI] [PubMed] [Google Scholar]
- 367.McCoy KL. Interaction between cannabinoid system and Toll-like receptors controls inflammation. Mediat Inflamm. 2016;2016:5831315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 368.Schwaeble W, Constantinescu CS. Relationship between cannabinoids and the immune system. Immunobiology. 2010;215(8):587 (Special Issue 8, 2010 Introduction). [DOI] [PubMed] [Google Scholar]
- 369.Tanasescu R, Constantinescu CS. Cannabinoids and the immune system: an overview. Immunobiology. 2010;215(8):588–97. [DOI] [PubMed] [Google Scholar]
- 370.Bayazit H, Selek S, Karababa IF, Cicek E, Aksoy N. Evaluation of oxidant/antioxidant status and cytokine levels in patients with cannabis use disorder. Clin Psychopharmacol Neurosci. 2017;15(3):237–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 371.Carlisle SJ, Marciano-Cabral F, Staab A, Ludwick C, Cabral GA. Differential expression of the CB2 cannabinoid receptor by rodent macrophages and macrophage-like cells in relation to cell activation. Int Immunopharmacol. 2002;2(1):69–82. [DOI] [PubMed] [Google Scholar]
- 372.Stella N. Cannabinoid and cannabinoid-like receptors in microglia, astrocytes, and astrocytomas. Glia. 2010;58(9):1017–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 373.Walter L, Franklin A, Witting A, Wade C, Xie Y, Kunos G, et al. Nonpsychotropic cannabinoid receptors regulate microglial cell migration. J Neurosci. 2003;23(4):1398–405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 374.Cutando L, Busquets-Garcia A, Puighermanal E, GomisGonzález M, Delgado-García JM, Gruart A, et al. Microglial activation underlies cerebellar deficits produced by repeated cannabis exposure. J Clin Investig. 2013;123(7):2816–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 375.Zamberletti E, Gabaglio M, Prini P, Rubino T, Parolaro D. Cortical neuroinflammation contributes to long-term cognitive dysfunctions following adolescent delta-9-tetrahydrocannabinol treatment in female rats. Eur Neuropsychopharmacol. 2015;25(12):2404–15. [DOI] [PubMed] [Google Scholar]
- 376.Lopez-Rodriguez AB, Llorente-Berzal A, Garcia-Segura LM, Viveros MP. Sex-dependent long-term effects of adolescent exposure to THC and/or MDMA on neuroinflammation and serotoninergic and cannabinoid systems in rats. Br J Pharmacol. 2014;171(6):1435–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 377.Zamberletti E, Gabaglio M, Grilli M, Prini P, Catanese A, Pittaluga A, et al. Long-term hippocampal glutamate synapse and astrocyte dysfunctions underlying the altered phenotype induced by adolescent THC treatment in male rats. Pharmacol Res. 2016;111:459–70. [DOI] [PubMed] [Google Scholar]
- 378.Valjent E, Maldonado R. A behavioural model to reveal place preference to delta 9-tetrahydrocannabinol in mice. Psychopharmacology (Berl). 2000;147(4):436–8. [DOI] [PubMed] [Google Scholar]
- 379.Lorenzetti V, Solowij N, Yücel M. The role of cannabinoids in neuroanatomic alterations in cannabis users. Biol Psychiatry. 2016;79(7):e17–31. [DOI] [PubMed] [Google Scholar]
- 380.Melis M, Frau R, Kalivas PW, Spencer S, Chioma V, Zamberletti E, et al. New vistas on cannabis use disorder. Neuropharmacology. 2017;124:62–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 381.Gray KM, Watson NL, Carpenter MJ, Larowe SD. N-Acetylcysteine (NAC) in young marijuana users: an open-label pilot study. Am J Addict. 2010;19(2):187–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 382.Gray KM, Carpenter MJ, Baker NL, DeSantis SM, Kryway E, Hartwell KJ, et al. A double-blind randomized controlled trial of N-acetylcysteine in cannabis-dependent adolescents. Am J Psychiatry. 2012;169(8):805–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 383.Gray KM, Sonne SC, McClure EA, Ghitza UE, Matthews AG, McRae-Clark AL, et al. A randomized placebo-controlled trial of N-acetylcysteine for cannabis use disorder in adults. Drug Alcohol Depend. 2017;177:249–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 384.Hutchinson MR, Northcutt AL, Chao LW, Kearney JJ, Zhang Y, Berkelhammer DL, Loram LC, Rozeske RR, Bland ST, Maier SF, Gleeson TT, Watkins LR. Minocycline suppresses morphineinduced respiratory depression, suppresses morphine-induced. Brain Behav Immun. 2008;22(8):1248–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 385.Zhang XQ, Cui Y, Cui Y, Chen Y, Na XD, Chen FY, Wei XH, Li YY, Liu XG, Xin WJ. Activation of p38 signaling in the microglia in the nucleus accumbens contributes to the acquisition and maintenance of morphine-induced conditioned place preference. Brain Behav Immun. 2012;26(2):318–25. [DOI] [PubMed] [Google Scholar]
- 386.Reissner KJ, Gipson CD, Tran PK, Knackstedt LA, Scofield MD, Kalivas PW. Glutamate transporter GLT-1 mediates N-acetylcysteine inhibition of cocaine reinstatement. Addict Biol. 2015;20(2):316–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 387.Miller WR, Fox RG, Stutz SJ, Lane SD, Denner L, Cunningham KA, Dineley KT. PPARγ agonism attenuates cocaine cue reactivity. Addict Biol. 2018;23(1):55–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 388.Sofuoglu M, Mooney M, Kosten T, Waters A, Hashimoto K. Minocycline attenuates subjective rewarding effects of dextroamphetamine in humans. Psychopharmacology (Berl). 2011;213(1):61–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 389.Amen SL, Piacentine LB, Ahmad ME, Li SJ, Mantsch JR, Risinger RC, Baker DA. Repeated N-acetyl cysteine reduces cocaine seeking in rodents and craving in cocaine-dependent humans. Neuropsychopharmacology. 2011;36(4):871–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 390.Alhaddad H, Das SC, Sari Y. Effects of ceftriaxone on ethanol intake: a possible role for xCT and GLT-1 isoforms modulation of glutamate levels in P rats. Psychopharmacology (Berl). 2014;231(20):4049–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 391.Das SC, Yamamoto BK, Hristov AM, Sari Y. Ceftriaxone attenuates ethanol drinking and restores extracellular glutamate concentration through normalization of GLT-1 in nucleus accumbens of male alcohol-preferring rats. Neuropharmacology. 2015;97:67–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 392.Rao PS, Sari Y. J Effects of ceftriaxone on chronic ethanol consumption: a potential role for xCT and GLT1 modulation of glutamate levels in male P rats. Mol Neurosci. 2014;54(1):71–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 393.Harris RA, Bajo M, Bell RL, Blednov YA, Varodayan FP, Truitt JM, de Guglielmo G, Lasek AW, Logrip ML, Vendruscolo LF, Roberts AJ, Roberts E, George O, Mayfield J, Billiar TR, Hackam DJ, Mayfield RD, Koob GF, Roberto M, Homanics GE. Genetic and Pharmacologic Manipulation of TLR4 Has Minimal Impact on Ethanol Consumption in Rodents. J Neurosci. 2017;37(5):1139–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 394.Schmaal L, Berk L, Hulstijn KP, Cousijn J, Wiers RW, van den Brink W. Efficacy of N-acetylcysteine in the treatment of nicotine dependence: a double-blind placebo-controlled pilot study. Eur Addict Res. 2011;17(4):211–6. [DOI] [PubMed] [Google Scholar]
