Next Article in Journal
Anoctamin 3: A Possible Link between Cluster Headache and Ca2+ Signaling
Next Article in Special Issue
Genetic Relationships Between Ethanol-Induced Conditioned Place Aversion and Other Ethanol Phenotypes in 15 Inbred Mouse Strains
Previous Article in Journal
Management of Sickle Cell Disease Pain among Adolescent and Pediatric Patients
Previous Article in Special Issue
Effects of Ethanol Exposure and Withdrawal on Neuronal Morphology in the Agranular Insular and Prelimbic Cortices: Relationship with Withdrawal-Related Structural Plasticity in the Nucleus Accumbens
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Metabotropic Glutamate Receptor Subtype 5 in Alcohol-Induced Negative Affect

by
Chelsea R. Kasten
,
Eleanor B. Holmgren
and
Tiffany A. Wills
*
LSU Health Sciences Center—New Orleans, Department of Cell Biology and Anatomy, Medical Education Building, 1901 Perdido Street, Room 6103, New Orleans, LA 70112, USA
*
Author to whom correspondence should be addressed.
Brain Sci. 2019, 9(8), 183; https://doi.org/10.3390/brainsci9080183
Submission received: 30 June 2019 / Revised: 26 July 2019 / Accepted: 28 July 2019 / Published: 30 July 2019

Abstract

:
Allosteric modulators of metabotropic glutamate 5 receptors (mGlu5 receptors) have been identified as a promising treatment to independently alleviate both negative affective states and ethanol-seeking and intake. However, these conditions are often comorbid and might precipitate one another. Acute and protracted ethanol withdrawal can lead to negative affective states. In turn, these states are primary drivers of alcohol relapse, particularly among women. The current review synthesizes preclinical studies that have observed the role of mGlu5 receptor modulation in negative affective states following ethanol exposure. The primary behavioral assays discussed are ethanol-seeking and intake, development and extinction of ethanol-associated cues and contexts, behavioral despair, and anxiety-like activity. The work done to-date supports mGlu5 receptor modulation as a promising target for mediating negative affective states to reduce ethanol intake or prevent relapse. Limitations in interpreting these data include the lack of models that use alcohol-dependent animals, limited use of adolescent and female subjects, and a lack of comprehensive evaluations of negative affective-like behavior.

1. Introduction

Metabotropic glutamate 5 receptors (mGlu5 receptors) represent a viable target for the treatment of alcohol-use disorders (AUDs) and negative affective phenotypes, and their use for each of these was recently independently reviewed [1,2,3]. However, negative affective states are also known to play a role in AUDs, as they are a primary driver of drinking and relapse behaviors [4]. “Negative affect” is a defined set of negative emotional states, which underlie many highly comorbid psychiatric disorders, including depression and anxiety [5]. Preclinically, negative affective symptoms can be clustered into changes in reinforcer seeking and consumption, behavioral despair, anxiety-like activity, increased threat monitoring, hypervigilance, and home cage activity [6,7,8]. These negative affective states might heighten sensitivity to alcohol-related cues and drive relapse [9,10]. Moreover, disorders that encompass negative affect are more prevalent in women and are highly comorbid with AUDs [11,12]. The current review will synthesize research that has investigated the role of mGlu5 receptors in alcohol use and negative affect, as well as identify gaps in the literature, particularly in regard to sex differences.
This review will primarily integrate the work involved in mGlu5 receptor modulation and its ability to regulate ethanol intake, the salience of ethanol-associated cues and contexts, and ethanol-induced behavioral despair and anxiety-like activity. mGlu5 and mGlu1 receptors comprise the group 1 metabotropic glutamate receptor class (mGlu1/5 receptors), which are Gαq/11-coupled receptors that regulate synaptic plasticity [13]. In these studies, mGlu5 receptors have been modulated using genetic manipulations or pharmacological tools. The role of global central nervous system knockout of mGlu5 receptors in drug use was first reported by Chiamulera et al. [14], who demonstrated that mGlu5-receptor-null mice do not acquire cocaine self-administration. Many tools beyond global knockout now exist, including cell-specific knockout [15,16], mGlu5 receptor deficiency [17], and mGlu5 receptor point mutations [18,19]. These studies used allosteric modulators, which are ligands that bind to a receptor site that is distinct from the endogenous or orthosteric ligand, as pharmacological interventions. Allosteric modulators negatively or positively regulate the activity of a receptor in the presence of its orthosteric ligand, but might also act as allosteric agonists or inverse agonists. mGlu5 receptor allosteric modulators discussed in the current review include the positive allosteric modulator (PAM), 3-Cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl)benzamide (CDPPB), and the negative allosteric modulators (NAM), 2-Methyl-6-(phenylethynyl)pyridine (MPEP) and 3-((2-Methyl-1,3-thiazol-4-yl)ethynyl)pyridine (MTEP). CDPPB has been noted for its interaction with the MPEP binding site, as well as its ability to mediate aberrant phenotypes associated with dysregulation of the ionotropic glutamate N-methyl-D-aspartate receptor (NMDAR) [20]. It should be noted that MPEP and MTEP both act as inverse agonists to block the constitutive activities of the mGlu5 receptors in vivo [21,22]. They also differ in their selectivity for mGlu5 receptors. Although MPEP is selective for mGlu5 receptors at lower doses, it becomes less selective as the dose increases and acts at other receptors, including NMDARs. The more recently developed MTEP conserves selectivity for mGlu5 receptors without demonstrating off target activity at NMDARs [2]. All of these drugs have been implicated as potential therapeutic treatments to alleviate negative affective symptomology and drug-seeking behavior [2,20].

2. Ethanol Intake

The ability of mGlu5 receptors to modulate ethanol intake has recently been extensively reviewed [3]. Generally, treatment with mGlu5 receptor NAMs results in decreased ethanol consumption and responding in 24 h access 2-bottle choice, limited access, and operant drinking paradigms (see Table 1). With the exception of Adams et al. [23] systemic mGlu5 receptor modulation consistently reduced drinking across a range of paradigms, species, and strains. Conversely, antagonism of mGlu1 receptors via systemic treatment of CPCCOEt was less effective at reducing ethanol intake or resulted in off-target effects, including reduced locomotion or sucrose intake [24,25,26]. Although higher doses of mGlu5 receptor NAMs also reduced locomotion and sucrose intake in some studies, these doses are beyond the efficacious dose for ethanol intake [26,27,28,29]. This further indicates that mGlu5 receptor-targeted treatment could be well tolerated as a clinical intervention compared to mGlu1-receptor pharmacological interventions. Finally, systemic MPEP prevents the alcohol deprivation effect following free-choice and operant ethanol access [24,30]. However, its efficacy may be reduced over repeated deprivation cycles [24], indicating the necessity of using alcohol-dependent models. Repeated alcohol exposure and withdrawal cycles promote neuroadaptations [9,10], which might lessen the efficacy of a drug that was initially promising.
Due to the allosteric properties of CDPPB, MPEP, and MTEP, it is important to consider whether alcohol exposure affects receptor availability and how that could inform the appropriate pharmacological treatment for different populations with AUDs. Using the highly potent and selective mGlu5 receptor NAM 18[F]-FPEB in PET scans, mGlu5 receptor availability has been demonstrated to be relatively stable in healthy humans over a 6 month period [38]. However, higher doses of MTEP are required to reduce ethanol consumption in alcohol-dependent rats [36], and alcohol has been demonstrated to alter mGlu5 receptor availability in both humans and rodents. In rodents, relatively low doses of forced ethanol over a two-week period enhances striatal, hippocampal, and cortical mGlu5 receptor availability compared to saline controls [39]. In contrast, chronic free-choice access to ethanol decreases mGlu5 receptor availability in the hippocampus and amygdala, when compared to baseline levels [40]. These PET studies lend support to the hypothesis that extensive alcohol exposure shifts the availability of mGlu5 receptors, thereby resulting in reduced efficacy of MTEP to reduce drinking in dependent rats [36]. Similar results have been found in humans, where increased mGlu5 receptor availability primarily in cortical regions is associated with “feeling high” during alcohol exposure in healthy, low-drinking humans [41]. Alcohol-dependent individuals have lower mGlu5 receptor availability compared to controls, across many striatal and cortical regions [42]. Availability of mGlu5 receptors recovers in a site- and time-dependent manner, across 6 months of alcohol abstinence, except in the hippocampus, accumbens, and thalamus [43]. This reduced mGlu5 receptor availability in alcohol-dependent subjects may be mediated by comorbid substance use, such as smoking status [44]. Non-smoking alcohol-dependent males show increased, not decreased, mGlu5 receptor availability in cortical regions and the amygdala at one month of abstinence [44]. Notably, reduced mGlu5 receptor availability is related to increased alcohol craving, regardless of smoking status [42,43,44]. Collectively, this work points toward dynamic regulation of mGlu5 receptor availability across early alcohol use, chronic alcohol use, alcohol abstinence, and comorbid drug use. It is still unclear from this work if there is a causal relationship between receptor availability and excessive ethanol drinking, dependence, and craving. Genetic variants in the mGluR-eEf2-α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) pathway, including GRM5, predict alcohol intake [45] and might independently regulate receptor availability. These findings complement the pharmacological studies mentioned above in pointing towards differences in mGlu5 receptor availability and pharmacological efficacy, depending on the type of alcohol exposure and genetic predisposition.
The studies detailed in Table 1 primarily focused on adult males; however, there is strong clinical evidence that supports the need to observe ethanol consumption in females and adolescents following mGlu5 receptor modulation. Adolescence is the time when alcohol use is typically initiated, and this use is known to be one of the strongest predictors for later development of AUDs [12,46]. While males are likely to use alcohol as positive reinforcement, females are more likely to use alcohol as a negative reinforcement coping mechanism [47]. Females are 2–3 times more likely to develop stress and anxiety disorders that might contribute to negative affective states, and this divergence of risk coincides with adolescent alcohol exposure [11]. It has been demonstrated that women have lower mGlu5 receptor availability across many brain regions [48], thereby indicating that there may be sex-differences in response to allosteric modulators. Cozzoli et al. [31] investigated the interaction of both adolescence and sex on ethanol intake. MTEP effectively reduced ethanol intake in both adolescent and adult male and female mice. However, during protracted abstinence (21 days), prior MTEP treatment showed no long-term effects on ethanol consumption in males of either age group. Females exposed to MTEP pretreatment during adolescence reduced their ethanol consumption in adulthood, whereas their adult-treated counterparts showed increased alcohol consumption. In another study, Parkitna et al. [15] demonstrated that knockdown of mGlu5 receptors on D1 neurons did not alter acquisition of ethanol intake under a continuous access instrumental response paradigm in females, but did inhibit an ethanol deprivation ramp-up during forced abstinence. Although males of the same strain demonstrated alcohol deprivation ramp-up of intake, it was not altered by mGlu5 receptor knockdown [16]. However, the male and female paradigms differed in length of alcohol history and instrumental response criteria, making it difficult to determine if the disparate findings were due to methodological- or sex-differences. Although mGlu5 receptor NAMs have not yet been investigated for their potential to alleviate AUD and comorbid symptomology, multiple treatments that target this system with minimal side effects have been developed for clinical use [49]. The preclinical studies indicate that alcohol duration, length of abstinence, age, and sex are all-important considerations in judging the effectiveness of mGlu5 receptor NAMs.

3. Ethanol-Associated Cues and Contexts

The role of mGlu5 receptors in learning and memory of discrete and contextual cues has been well documented. mGlu5 receptor activity contributes to neural plasticity via both long-term depression (LTD), as well as long-term potentiation (LTP) via mGlu5-NMDAR interactions discussed in Section 6 [13,50]. In rodent models of non-dependent ethanol intake, mGlu5 receptor modulation effectively reduces the salience of ethanol-associated cues and contexts when administered following the cue–ethanol association (see Table 2). The ability to modulate the salience of ethanol-associated cues and contexts, plays an important role in reducing susceptibility to relapse, making it a critical target for pharmacological intervention [51,52]. Two paradigms have been primarily used to observe the role of mGlu5 receptors in ethanol-associated cues and contexts—ethanol cue-induced reinstatement and conditioned place preference (CPP).
With the exception of Adams et al. [23,57], systemic and site-specific negative allosteric modulation of mGlu5 receptors was found to reduce cue-induced reinstatement in the presence of discrete cues, such as a light cue [53,54,55], or diffuse contextual stimuli, such as an olfactory scent [30]. Contextual cues have been posited as more analogous to cues that induce drug craving and seeking in humans than discrete cues, due to their role in indicating general drug availability, transfer of salience, and the difficulty involved in extinguishing these cues [52]. Although Adams and colleagues [23,57] partially contributed their null findings to the low dose of MTEP used, it is important to note that their cue-induced reinstatement paradigm utilized a contextual scent to signal ethanol availability and inbred, high-alcohol-preferring iP rats. Using a discrete-cue paradigm, mGlu5 receptor modulation was found to readily block cue-induced reinstatement at a relatively low dose in iP rats [55]. Therefore, it might be speculated that a genetic predisposition for alcohol preference makes animals resilient to pharmacological intervention, to reduce particularly salient ethanol-associated cues. Without the investigation of higher drug doses in the iP rats, it cannot be concluded whether these disparate results were due to less sensitivity to mGlu5 receptor intervention under contextual cue paradigms, or whether mGlu5 receptors only play a role in discrete-cued reinstatement when there is a genetic predisposition to consume ethanol.
CPP, which quantifies the reinforcing value of ethanol by observing the amount of time spent in an ethanol-paired context, can be broken down into the cue/context learning phase (acquisition) and the expression of the learned association [52]. Pharmacologically or genetically reducing the activity of mGlu5 receptors results in impaired ethanol CPP, indicating that mGlu5 receptor activity contributes to the associating contexts, with ethanol. Notably, this effect appears to be restricted to the expression [25,56,58] but not acquisition of CPP [56,59]. In the acquisition studies, drug was administered prior to ethanol during the contextual-pairing sessions, whereas drug was administered without any ethanol on board during the expression test sessions. This might indicate that mGlu5 receptor modulation is not able to overcome the salience of ethanol exposure as it occurs, but rather it blocks the recall of ethanol-associated cues.
In direct opposition to the current studies, it has been reported that mGlu5 receptor NAMs often inhibit the extinction of contextual and spatial memories, whereas positive modulation enhances extinction [60,61,62,63,64]. Notably, these studies consist primarily of aversive learning conditions, such as avoidance learning, startle response, and fear conditioning. Similar to the currently discussed findings on ethanol context and cues, negative mGlu5 receptor modulation has been shown to block context-paired locomotor conditioning to cocaine and methamphetamine CPP [65,66]. Acknowledging the divergent effects of negative mGlu5 receptor modulation on cues and contexts associated with positive versus negative stimuli is important. In states of dependence and withdrawal, positive ethanol-associated cues might transfer their association to negative affective states. Therefore, these once positive cues might be more similar to the aversive cues that are enhanced by negative mGlu5 receptor modulation. Sidhpura et al. [36] demonstrated that, although MTEP was still effective at blocking stress-induced reinstatement in dependent rats, it was more effective in non-dependent rats. mGlu5 receptor NAMs might also be an insufficient treatment for people experiencing an ethanol relapse. Positive mGlu5 modulation has been demonstrated to rescue impaired spatial learning following heavy ethanol exposure [67], indicating its safety and efficacy in models of dependence. Although the studies discussed within this section favor negative mGlu5 receptor modulation for mediating ethanol-associated cues and contexts, positive mGlu5 receptor modulation might be a better course of treatment under aversive states associated with alcohol withdrawal and relapse.
Females were not included in any of the discussed studies that observed cue-induced cue reinstatement. In rodents, acute pharmacological stress significantly enhances cue-induced ethanol reinstatement in females, but not males [68]. In humans, stress has not been demonstrated to enhance ethanol craving or relapse to a greater degree in females than males. However, these human studies either did not report estradiol levels or estrous status [69,70], or restricted female testing to periods when circulating estradiol levels were low [71]. In rodents, circulating estradiol levels were significantly positively correlated with the magnitude of stress-induced reinstatement [68]. Further, females demonstrate an enhanced ethanol CPP that is dependent upon circulating hormones [72], as well as alterations in drug efficacy to reduce ethanol intake based on estrous status [73]. These data indicate that circulating hormones mediate the salience of cues and stress on ethanol-associated activity and should be included in human female studies. There is support for direct interaction of mGlu1/5 receptors and estrogen receptors (ER) signaling (see Section 6). This convergence of signaling cascades might indicate that mGlu5 receptor modulation would be a particularly salient treatment in female populations.

4. Behavioral Despair

Behavioral despair, or anhedonic activity, is observed in animal models that represent depressive-like behavior. Although many animal models of behavioral despair exist—including reduction in intake of appetitive reinforcers, the forced swim test, the tail suspension test, social interaction, and response to novelty [8,74]—few have been observed following ethanol administration and/or mGlu5 receptor manipulation (see Table 3). Of these studies, the forced swim task has been the predominantly used paradigm. This task observes the time spent immobile in a container of water. Time spent immobile is decreased by treatment with antidepressants, indicating translational relevance [74]. Limited access ethanol in adult male mice reliably induces behavioral despair in the forced swim test 24 h into withdrawal [75,76,77]. This effect is rescued by systemic MTEP treatment, but not site-specific treatment targeting the NAc shell [75,76]. Conversely, systemic treatment with the mGlu5 receptor PAM, CDPPB, exacerbates the effect of ethanol withdrawal on behavioral despair [75]. Adolescent ethanol exposure, however, does not result in a behavioral despair phenotype in male mice 24 h into withdrawal. In contrast, CDPPB administration is able to induce a behavioral despair phenotype in water drinking controls [75]. Interestingly, protracted withdrawal from adolescent alcohol does result in a behavioral despair phenotype [76], but it is not known if it can be rescued via systemic mGlu5 receptor modulation.
Although mGlu5 receptors appear to be a promising target for rescuing behavioral despair induced by ethanol exposure, these studies suffer from lack of diversity in tests, ethanol exposure paradigms, sex, and age. All studies discussed in this section used a 14-day drinking-in-the-dark exposure in male B6 mice. No studies have observed the effects of ethanol dependence, prolonged ethanol exposure, or protracted ethanol withdrawal in adulthood on behavioral despair. These studies are necessary to accurately capture the ability of mGlu5 receptor modulation to mediate the negative-affective withdrawal phenotype that promotes relapse susceptibility. Male and female rodents also express behavioral despair in a sex-dependent manner. For example, females are susceptible to the forced swim test, but relatively resilient to psychosocial models of despair [74]. The forced swim task suffers from many criticisms, which include the lack of translational value for treatment development, dependence on physical activity, and differing survival strategies to conserve energy [74,78]. However, the forced swim test is high-throughput, and engages overlapping neural circuitry with humans suffering from depression [74], making it a valuable tool when paired with other behavioral despair tests. Complementary tasks may include seeking and consumption of non-drug reinforcers, tail suspension task, social interaction, response to novelty, and observation of normative home-cage activities such as grooming [6,8]. As these tests result in sexually distinct phenotypes that are not consistent between tasks [8], it is important to include multiple behavioral paradigms to observe how ethanol alters the complete behavioral despair phenotype. Finally, as mGlu5 receptor modulation shows promise in these studies, it should be examined at both younger and older ages, following alcohol exposure, dependence, and protracted withdrawals. Depression at both young and old age is associated with poor outcomes and limited response to traditional antidepressants [74], making the mGlu5 receptors a promising target.
mGlu5 receptors have been extensively implicated in major depression disorder (MDD) at the clinical and preclinical levels, as recently reviewed by Esterlis et al. [79]. Similar to the studies discussed that have observed mGlu5 receptor availability in alcohol use disorders (see Section 2), studies observing those with MDD without alcohol and substance use disorders have reported mixed findings. One study has reported increased post-mortem Grm5 expression in the locus coeruleus of MDD individuals, noting the important role of locus coeruleus excitability in MDD [80]. Studies reporting reduced mGlu5 receptor availability in those with MDD have been conducted in non-smoking populations [81,82], whereas those that reported no differences overwhelmingly included smoking individuals [83,84,85,86]. Smoking status also mediates the relationship between mGlu5 receptor availability and alcohol use, but it appears that smoking is responsible for the reduced mGlu5 receptor availability in heavy drinkers [44]. Although the relationships between mGlu5 receptor availability and smoking status in heavy alcohol use and MDD are divergent, it is still notable that smoking status might alter response to mGlu5 receptor modulators as behavioral treatments in each of these populations. Ketamine, which was recently approved by the FDA for treatment-resistant MDD, rapidly reduces availability of mGlu5 receptors in non-smokers with MDD and in healthy controls. The magnitude of reduction of receptor availability in the hippocampus was positively correlated with a reduction in symptoms of depression [81]. This relationship between mGlu5 receptor availability and depression symptomology has also been reported in non-smoking individuals not treated with ketamine [82]. Although the effects of ketamine have been linked to mGlu5 receptors, directly targeting mGlu5 receptors with NAMs has not been demonstrated to treat symptoms of MDD in clinical trials [87,88]. Notably, the primary outcome of these studies was the Montgomery–Åsberg Depression Rating Scale (MADRS), which is clinician-rated during an interview session. However, when patients are asked to self-report, negative mGlu5 receptor modulation significantly improves depressive symptomology and quality of life when paired with a traditional antidepressant [88]. These results indicate that treatment with mGlu5 receptor modulators might alleviate internal feelings of depressive symptomology that promote excessive alcohol intake.

5. Anxiety-Like Activity

Anxiety-like activity is a major component of negative affective behavior, as well as a primary driver of stress and stress-induced relapse [5,10]. Several studies have observed the ability of mGlu5 receptor modulation to alter alcohol-induced unconditioned anxiety-like behavior across a range of paradigms. These paradigms include approach–avoidance conflict tasks (elevated plus maze, light/dark box, and the open field task) [89], as well as the marble burying task. Although marble burying is poorly correlated with traditional measures of anxiety, it is regarded as a perseverative, investigative activity that can be pharmacologically manipulated [90,91]. With few exceptions [58,75], all papers detailed in Table 4 observed increases in anxiety-like activity following ethanol exposure, which were overwhelmingly rescued by mGlu5 receptor NAM administration.
The efficacy of mGlu5 receptor NAMs to reduce heightened anxiety-like activity is consistent across ethanol i.p. administration [92], ethanol liquid diet [93], and free-choice limited ethanol access [75,76]. Within adult animals, the findings were also consistent across behavioral assays. This is notable due to the poor predictive validity of each of these tests on their own [89]. Further, in the absence of alcohol, mGlu5 receptor modulation is a promising target for treatment of anxiety disorders. A majority of reports using mGlu5 receptor modulation to alter anxiety-like activity report anxiolytic responses, whereas serotonergic, endocannabinoid, neuropeptide, and other glutamatergic targets often report inactivity of the tested compounds, or even anxiogenic activity [94,95]. In these studies, mGlu5 receptor modulation had minimal effects on anxiety-like activity in control mice, contrary to its predominately anxiolytic profile in many assays [95]. One reason for this might be that these tests employed parameters that evoked low baseline anxiety levels (such as low light intensity) to be able to detect heightened anxiety-like activity present in alcohol exposed mice. In typical anxiety-like assays, ceiling levels of anxiety are often provoked by bright lights, aversive or threatening stimuli, or conflict [95]. As such, the current studies indicate that targeting mGlu5 receptors might normalize maladaptive behavior that is present following alcohol use, without disrupting normal system function.
Although the studies examining anxiety-like activity use a wide range of ethanol exposures and behavioral outcomes, they still suffer from limitations of age and sex, with adult males being the primary demographic studied. With the exception of Lee et al. [75], mGlu5 receptor modulation was able to rescue anxiety-like phenotypes observed within 48 h of the last ethanol vapor. Lee et al. [75] were unable to demonstrate an enhanced anxiety-like profile in adolescent males following ethanol exposure. However, it has been well-documented that alcohol exposure during adolescence kindles anxiety-like behavior during protracted withdrawal, as mice age into adulthood [75,96,97,98,99,100,101]. In the context of preventing negative-affect-induced relapse, it is necessary to investigate whether mGlu5 receptor modulation might also rescue enhanced anxiety-like activity that occurs during extensive ethanol abstinence. Similarly, females may be especially sensitive to anxiety during periods of abstinence [11,47], with protracted withdrawal from adolescent alcohol resulting in enhanced anxiety-like and despair behavior [99,102]. These results highlight the need to observe the ability of mGlu5 receptor modulation to alter anxiety-like activity in males and females during protracted withdrawal from alcohol.

6. Synaptic Plasticity

While the studies discussed up to this point have focused on the behavioral outcomes of mGlu1/5 receptor activation, much is also known about the impacts of the cellular mechanisms of these receptors by drugs of abuse and negative affect. mGlu1/5 receptors are located postsynaptically or perisynaptically and are anchored to the postsynaptic density by interactions with Homer and SHANK. While mGlu1/5 receptors are key regulators of excitatory synaptic plasticity through both LTD and LTP, the following discussion focuses on mGlu1/5 regulation of LTD. Generally in regions like the bed nucleus of the stria terminalis (BNST) and striatum, mGlu1/5 receptor activation leads to phospholipase C (PLC) enhancement of PIP2, which in turn activates two divergent downstream pathways. One involves the activation of IP3 pathway and release of endoplasmic reticulum Ca2+ subsequent protein kinase C (PKC), mitogen-activated protein kinase kinase (MEK), and Erk1/2 activation, which can ultimately activate Arc. Secondly, activation of the IP3 pathway produces diacyl-glycerol (DAG), which is then acted upon by PLC and DAG lipase (DAGL) to produce the endocannabinoid, 2-AG. In LTD, the initial (early) phase of this LTD is initiated by generation of 2-AG, which is released from the postsynaptic neuron and activates presynaptic type 1 cannabinoid receptors (CB1 receptor). The activation of presynaptic CB1 receptors produces a reduction in glutamate release or an enhancement of GABA release. The maintenance (late) phase of this LTD is initiated through the actions of the IP3 pathway (discussed above) that ultimately produces the internalization of AMPARs [13]. The reliance on a raise in postsynaptic Ca2+ and subsequent activation of PKC or PLC, as well as the involvement of an endocannabinoid signaling, differ by brain region (reviewed in [13]). Additionally, the mode of LTD induction [drug induced via (S)-3, 5-dihydroxyphenylglycine (DHPG), paired-pulse-induces, or frequency-induced] is also thought to influence the reliance on certain mechanisms (reviwed in [13]). Further the anatomical contributions of mGlu1 receptors versus mGlu5 receptors differ by brain region.
The extended amygdala is a collection of brain structures including the nucleus accumbens shell (NAc shell), the bed nucleus of the stria terminalis (BNST), and the central nucleus of the amygdala (CeA). These brain structures are known to play critical roles in in the modulation of negative affect and stress, particularly in the context of withdrawal [4]. In the BNST of male mice, mGlu1/5 receptor-mediated LTD is disrupted by chronic cocaine during both acute withdrawal and prolonged abstinence [103,104]. This cocaine-induced mGlu1/5 receptor-mediated LTD disruption is manifested by internalization of GluA2-containing AMPARs (calcium-impermeable), followed by replacement with calcium-permeable-AMPARs in the ventral tegmental area (VTA) and NAc [105,106,107,108]. Outside the extended amygdala, a similar disruption of mGlu1/5 receptor-mediated LTD is also found in the hippocampus during acute withdrawal from chronic ethanol vapor in male mice. In the CeA, there is a role for mGlu1/5-Homer signaling on ethanol binge-drinking [34]. A large body of literature from the Szumlinski lab finds that mGlu1/5 receptor signaling effects on ethanol are mediated through the interaction with Homer 2 [18,34,109,110,111,112,113]. Recently, this same group expanded on this work to find that Erk phosphorylation enhances mGlu5-Homer interaction in the BNST and this action attenuates ethanol drinking [19].
This mGlu5 receptor signaling mechanism was also found to be critical for estradiol-driven potentiation of psychostimulant-induced behaviors in female rodents [114,115]. Estradiol activates ERα through activation of mGlu1/5 receptors, thereby activating CREB and phosphorylated PLC through MAPK, independent of glutamate activation. Estradiol-induced CREB phosphorylation is differentially mediated depending on the brain region. mGlu5 receptor-dependent regions include the dorsal striatum and NAc core, whereas the NAc shell is mGlu1a receptor-dependent. Estradiol’s interactions with mGlu1/5 receptors also site-specifically alters brain morphology, decreasing dendritic spines in the NAc core while increasing dendritic spines in the NAc shell. Rodent models of chronic cocaine use have demonstrated that females have fast acquisition, enhanced escalation, and greater reinstatement. ER/mGlu receptor signaling is thought to be responsible for a majority of the sex differences in cocaine behaviors and the neural transmission cocaine phenotypes elicited in females [116]. Given the efficacy of mGlu5 receptor modulation in males and the role of female sex hormones contributing to these behaviors in the cocaine literature, it might be expected that females would demonstrate enhanced ethanol intake, stronger associations with ethanol-associated cues and contexts, and enhanced behavioral despair and anxiety that would be particularly responsive to mGlu5 receptor modulation.

7. Conclusions

The data currently reviewed indicate that mGlu5 receptor modulation is a promising target for negative affect-like behavior associated with alcohol use disorders. mGlu5 receptor modulation is able to reduce ethanol intake, salience of ethanol-associated cues and contexts, behavioral despair, and anxiety-like activity. In humans, alcohol consumption manifests in many ways. This includes no alcohol use, light and recreational use, and dangerous levels of binging and intoxication. Currently, the literature suggests that mGlu5 receptors contribute to sex-specific neuroadaptations following alcohol use. These adaptations appear to be dependent upon age of onset of use, frequency of use, length of use, and length of abstinence from ethanol. Although the current studies touch on these points, the field is ripe for investigation of sex-differences, adolescent alcohol exposure, the role of alcohol dependence, and the effect of varying periods of withdrawal on the interaction of negative affect with alcohol intake and seeking. In particular, females and those exposed to alcohol during adolescence might be particularly susceptible to developing these negative affective states following protracted withdrawal from ethanol due to the role of developmental sex hormones in neuroadaptations underlying mGlu5 receptor signaling. The current studies also primarily focused on negative affective states following ethanol exposure. However, negative affective states often precipitate relapse. Therefore, future studies are needed to observe whether mGlu5 receptor modulation during periods of negative affect, such as chronic or unpredictable stressors, might work to alleviate ethanol intake. Finally, studies should consider using more than one task to observe behavioral despair and anxiety-like activity, as these phenotypes might manifest differently based on sex, age of exposure, and length of exposure or withdrawal. Although further work is required to broadly ensure the safety and efficacy of mGlu5 receptor modulation, the current work supports this system as a promising target for treating both ethanol-induced negative affect, as well as preventing negative affect-induced relapse.

Author Contributions

Conceptualization: C.R.K., E.B.H., and T.A.W.; Methodology: C.R.K. and T.A.W.; Investigation: C.R.K., E.B.H., and T.A.W.; Writing—Original Draft Preparation: C.R.K.; Writing—Review & Editing: C.R.K., E.B.H., and T.A.W.; Funding Acquisition: T.A.W.

Funding

This work was supported by NIAAA grant AA022651 (TAW) and The Biomedical Alcohol Research Training Program (NIAAA funded T32: AA007577).

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Peterlik, D.; Flor, P.J.; Uschold-Schmidt, N. The Emerging Role of Metabotropic Glutamate Receptors in the Pathophysiology of Chronic Stress-Related Disorders. Curr. Neuropharmacol. 2016, 14, 514–539. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Olive, M.F. Metabotropic glutamate receptor ligands as potential therapeutics for addiction. Curr. Drug Abuse Rev. 2009, 2, 83–98. [Google Scholar] [CrossRef] [PubMed]
  3. Goodwani, S.; Saternos, H.; Alasmari, F.; Sari, Y. Metabotropic and ionotropic glutamate receptors as potential targets for the treatment of alcohol use disorder. Neurosci. Biobehav. Rev. 2017, 77, 14–31. [Google Scholar] [CrossRef] [PubMed]
  4. Koob, G.F.; Volkow, N.D. Neurocircuitry of Addiction. Neuropsychopharmacology 2010, 35, 217–238. [Google Scholar] [CrossRef] [PubMed]
  5. Brown, T.A.; Barlow, D.H. A proposal for a dimensional classification system based on the shared features of the DSM-IV anxiety and mood disorders: Implications for assessment and treatment. Psychol. Assess. 2009, 21, 256–271. [Google Scholar] [CrossRef] [PubMed]
  6. Wright, J.S.; Panksepp, J. Toward affective circuit-based preclinical models of depression: Sensitizing dorsal PAG arousal leads to sustained suppression of positive affect in rats. Neurosci. Biobehav. Rev. 2011, 35, 1902–1915. [Google Scholar] [CrossRef] [PubMed]
  7. Kirlic, N.; Aupperle, R.L.; Rhudy, J.L.; Misaki, M.; Kuplicki, R.; Sutton, A.; Alvarez, R.P. Latent variable analysis of negative affect and its contributions to neural responses during shock anticipation. Neuropsychopharmacology 2019, 44, 695–702. [Google Scholar] [CrossRef] [PubMed]
  8. Goodwill, H.L.; Manzano-Nieves, G.; Gallo, M.; Lee, H.-I.; Oyerinde, E.; Serre, T.; Bath, K.G. Early life stress leads to sex differences in development of depressive-like outcomes in a mouse model. Neuropsychopharmacology 2019, 44, 711–720. [Google Scholar] [CrossRef]
  9. Becker, H.C. Alcohol dependence, withdrawal, and relapse. Alcohol Res. Health 2008, 31, 348–361. [Google Scholar]
  10. Becker, H.C. Influence of stress associated with chronic alcohol exposure on drinking. Neuropharmacology 2017, 122, 115–126. [Google Scholar] [CrossRef]
  11. Craske, M.G.; Stein, M.B.; Eley, T.C.; Milad, M.R.; Holmes, A.; Rapee, R.M.; Wittchen, H.-U. Anxiety disorders. Nat. Rev. Dis. Prim. 2017, 3, 17024. [Google Scholar] [CrossRef] [PubMed]
  12. McHugh, R.K.; Votaw, V.R.; Sugarman, D.E.; Greenfield, S.F. Sex and gender differences in substance use disorders. Clin. Psychol. Rev. 2018, 66, 12–23. [Google Scholar] [CrossRef] [PubMed]
  13. Lüscher, C.; Huber, K.M. Group 1 mGluR-dependent synaptic long-term depression: Mechanisms and implications for circuitry and disease. Neuron 2010, 65, 445–459. [Google Scholar] [CrossRef] [PubMed]
  14. Chiamulera, C.; Epping-Jordan, M.P.; Zocchi, A.; Marcon, C.; Cottiny, C.; Tacconi, S.; Corsi, M.; Orzi, F.; Conquet, F. Reinforcing and locomotor stimulant effects of cocaine are absent in mGluR5 null mutant mice. Nat. Neurosci. 2001, 4, 873–874. [Google Scholar] [CrossRef] [PubMed]
  15. Parkitna, J.R.; Sikora, M.; Gołda, S.; Gołembiowska, K.; Bystrowska, B.; Engblom, D.; Bilbao, A.; Przewlocki, R. Novelty-Seeking Behaviors and the Escalation of Alcohol Drinking After Abstinence in Mice Are Controlled by Metabotropic Glutamate Receptor 5 on Neurons Expressing Dopamine D1 Receptors. Biol. Psychiatry 2013, 73, 263–270. [Google Scholar] [CrossRef] [PubMed]
  16. Eisenhardt, M.; Leixner, S.; Spanagel, R.; Bilbao, A. Quantification of alcohol drinking patterns in mice. Addict. Biol. 2015, 20, 1001–1011. [Google Scholar] [CrossRef]
  17. Bird, M.K.; Kirchhoff, J.; Djouma, E.; Lawrence, A.J. Metabotropic glutamate 5 receptors regulate sensitivity to ethanol in mice. Int. J. Neuropsychopharmacol. 2008, 11, 765–774. [Google Scholar] [CrossRef] [Green Version]
  18. Cozzoli, D.K.; Goulding, S.P.; Zhang, P.W.; Xiao, B.; Hu, J.-H.; Ary, A.W.; Obara, I.; Rahn, A.; Abou-Ziab, H.; Tyrrel, B.; et al. Binge drinking upregulates accumbens mGluR5-Homer2-PI3K signaling: Functional implications for alcoholism. J. Neurosci. 2009, 29, 8655–8668. [Google Scholar] [CrossRef]
  19. Campbell, R.R.; Domingo, R.D.; Williams, A.R.; Wroten, M.G.; McGregor, H.A.; Waltermire, R.S.; Greentree, D.I.; Goulding, S.P.; Thompson, A.B.; Lee, K.M.; et al. Increased Alcohol-Drinking Induced by Manipulations of mGlu5 Phosphorylation within the Bed Nucleus of the Stria Terminalis. J. Neurosci. 2019, 39, 2745–2761. [Google Scholar] [CrossRef] [Green Version]
  20. Stauffer, S.R. Progress toward positive allosteric modulators of the metabotropic glutamate receptor subtype 5 (mGluR5). ACS Chem. Neurosci. 2011, 2, 450–470. [Google Scholar] [CrossRef]
  21. Pagano, A.; Ruegg, D.; Litschig, S.; Stoehr, N.; Stierlin, C.; Heinrich, M.; Floersheim, P.; Prezèau, L.; Carroll, F.; Pin, J.P.; et al. The non-competitive antagonists 2-methyl-6-(phenylethynyl)pyridine and 7-hydroxyiminocyclopropan[b]chromen-1a-carboxylic acid ethyl ester interact with overlapping binding pockets in the transmembrane region of group I metabotropic glutamate receptors. J. Biol. Chem. 2000, 275, 33750–33758. [Google Scholar] [CrossRef] [PubMed]
  22. Gould, R.W.; Amato, R.J.; Bubser, M.; Joffe, M.E.; Nedelcovych, M.T.; Thompson, A.D.; Nickols, H.H.; Yuh, J.P.; Zhan, X.; Felts, A.S.; et al. Partial mGlu5 Negative Allosteric Modulators Attenuate Cocaine-Mediated Behaviors and Lack Psychotomimetic-Like Effects. Neuropsychopharmacology 2016, 41, 1166–1178. [Google Scholar] [CrossRef] [PubMed]
  23. Adams, C.L.; Short, J.L.; Lawrence, A.J. Cue-conditioned alcohol seeking in rats following abstinence: Involvement of metabotropic glutamate 5 receptors. Br. J. Pharmacol. 2010, 159, 534–542. [Google Scholar] [CrossRef] [PubMed]
  24. Schroeder, J.P.; Overstreet, D.H.; Hodge, C.W. The mGluR5 antagonist MPEP decreases operant ethanol self-administration during maintenance and after repeated alcohol deprivations in alcohol-preferring (P) rats. Psychopharmacology 2005, 179, 262–270. [Google Scholar] [CrossRef] [PubMed]
  25. Lominac, K.D.; Kapasova, Z.; Hannun, R.A.; Patterson, C.; Middaugh, L.D.; Szumlinski, K.K. Behavioral and neurochemical interactions between Group 1 mGluR antagonists and ethanol: Potential insight into their anti-addictive properties. Drug Alcohol Depend. 2006, 85, 142–156. [Google Scholar] [CrossRef] [PubMed]
  26. Hodge, C.W.; Miles, M.F.; Sharko, A.C.; Stevenson, R.A.; Hillmann, J.R.; Lepoutre, V.; Besheer, J.; Schroeder, J.P. The mGluR5 antagonist MPEP selectively inhibits the onset and maintenance of ethanol self-administration in C57BL/6J mice. Psychopharmacology 2006, 183, 429–438. [Google Scholar] [CrossRef] [PubMed]
  27. Cowen, M.S.; Djouma, E.; Lawrence, A.J. The metabotropic glutamate 5 receptor antagonist 3-[(2-methyl-1,3-thiazol-4-yl)ethynyl]-pyridine reduces ethanol self-administration in multiple strains of alcohol-preferring rats and regulates olfactory glutamatergic systems. J. Pharmacol. Exp. Ther. 2005, 315, 590–600. [Google Scholar] [CrossRef]
  28. Cowen, M.S.; Krstew, E.; Lawrence, A.J. Assessing appetitive and consummatory phases of ethanol self-administration in C57BL/6J mice under operant conditions: Regulation by mGlu5 receptor antagonism. Psychopharmacology 2007, 190, 21–29. [Google Scholar] [CrossRef]
  29. Besheer, J.; Faccidomo, S.; Grondin, J.J.M.; Hodge, C.W. Regulation of motivation to self-administer ethanol by mGluR5 in alcohol-preferring (P) rats. Alcohol. Clin. Exp. Res. 2008, 32, 209–221. [Google Scholar] [CrossRef]
  30. Bäckström, P.; Bachteler, D.; Koch, S.; Hyytiä, P.; Spanagel, R. mGluR5 Antagonist MPEP Reduces Ethanol-Seeking and Relapse Behavior. Neuropsychopharmacology 2004, 29, 921–928. [Google Scholar] [CrossRef]
  31. Cozzoli, D.K.; Strong-Kaufman, M.N.; Tanchuck, M.A.; Hashimoto, J.G.; Wiren, K.M.; Finn, D.A. The effect of mGluR5 antagonism during binge drinking on subsequent ethanol intake in C57BL/6J mice: Sex- and age-induced differences. Alcohol. Clin. Exp. Res. 2014, 38, 730–738. [Google Scholar] [CrossRef] [PubMed]
  32. Blednov, Y.A.; Adron Harris, R. Metabotropic glutamate receptor 5 (mGluR5) regulation of ethanol sedation, dependence and consumption: Relationship to acamprosate actions. Int. J. Neuropsychopharmacol. 2008, 11, 775–793. [Google Scholar] [CrossRef] [PubMed]
  33. McMillen, B.A.; Crawford, M.S.; Kulers, C.M.; Williams, H.L. Effects of a metabollic, mGlu5, glutamate receptor antagonist on ethanol consumption by genetic drinking rats. Alcohol Alcohol. 2005, 40, 494–497. [Google Scholar] [CrossRef] [PubMed]
  34. Cozzoli, D.K.; Courson, J.; Wroten, M.G.; Greentree, D.I.; Lum, E.N.; Campbell, R.R.; Thompson, A.B.; Maliniak, D.; Worley, P.F.; Jonquieres, G.; et al. Binge alcohol drinking by mice requires intact group1 metabotropic glutamate receptor signaling within the Central nucleus of the Amygdale. Neuropsychopharmacology 2014, 39, 435–444. [Google Scholar] [CrossRef] [PubMed]
  35. Gass, J.T.; Olive, M.F. Role of protein kinase C epsilon (PKCvarepsilon) in the reduction of ethanol reinforcement due to mGluR5 antagonism in the nucleus accumbens shell. Psychopharmacology 2009, 204, 587–597. [Google Scholar] [CrossRef] [PubMed]
  36. Sidhpura, N.; Weiss, F.; Martin-Fardon, R. Effects of the mGlu2/3 Agonist LY379268 and the mGlu5 Antagonist MTEP on Ethanol Seeking and Reinforcement Are Differentially Altered in Rats with a History of Ethanol Dependence. Biol. Psychiatry 2010, 67, 804–811. [Google Scholar] [CrossRef] [Green Version]
  37. Besheer, J.; Grondin, J.J.M.; Cannady, R.; Sharko, A.C.; Faccidomo, S.; Hodge, C.W. Metabotropic Glutamate Receptor 5 Activity in the Nucleus Accumbens Is Required for the Maintenance of Ethanol Self-Administration in a Rat Genetic Model of High Alcohol Intake. Biol. Psychiatry 2010, 67, 812–822. [Google Scholar] [CrossRef] [Green Version]
  38. Leurquin-Sterk, G.; Postnov, A.; de Laat, B.; Casteels, C.; Celen, S.; Crunelle, C.L.; Bormans, G.; Koole, M.; Van Laere, K. Kinetic modeling and long-term test-retest reproducibility of the mGluR5 PET tracer 18F-FPEB in human brain. Synapse 2016, 70, 153–162. [Google Scholar] [CrossRef]
  39. Nandi, A.; Valentine, H.; McCaul, M.; Wong, D. Glutamatergic abnormalities in a rodent model of alcohol abuse. J. Nucl. Med. 2016, 57, 1866a. [Google Scholar]
  40. de Laat, B.; Weerasekera, A.; Leurquin-Sterk, G.; Gsell, W.; Bormans, G.; Himmelreich, U.; Casteels, C.; Van Laere, K. Effects of alcohol exposure on the glutamatergic system: A combined longitudinal 18F-FPEB and 1H-MRS study in rats. Addict. Biol. 2019, 24, 696–706. [Google Scholar] [CrossRef]
  41. Leurquin-Sterk, G.; Ceccarini, J.; Crunelle, C.L.; Weerasekera, A.; de Laat, B.; Himmelreich, U.; Bormans, G.; Van Laere, K. Cerebral dopaminergic and glutamatergic transmission relate to different subjective responses of acute alcohol intake: An in vivo multimodal imaging study. Addict. Biol. 2018, 23, 931–944. [Google Scholar] [CrossRef] [PubMed]
  42. Leurquin-Sterk, G.; Ceccarini, J.; Crunelle, C.L.; de Laat, B.; Verbeek, J.; Deman, S.; Neels, H.; Bormans, G.; Peuskens, H.; Van Laere, K. Lower Limbic Metabotropic Glutamate Receptor 5 Availability in Alcohol Dependence. J. Nucl. Med. 2018, 59, 682–690. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Ceccarini, J.; Leurquin-Sterk, G.; Crunelle, C.; De Laat, B.; Bormans, G.; Peuskens, H.; Van Laere, K. Recovery of decreased metabotropic glutamate receptor 5 availability in abstinent alcohol-dependent subjects. J. Nucl. Med. 2017, 58, 14. [Google Scholar]
  44. Akkus, F.; Mihov, Y.; Treyer, V.; Ametamey, S.M.; Johayem, A.; Senn, S.; Rösner, S.; Buck, A.; Hasler, G. Metabotropic glutamate receptor 5 binding in male patients with alcohol use disorder. Transl. Psychiatry 2018, 8, 17. [Google Scholar] [CrossRef] [PubMed]
  45. Meyers, J.L.; Salling, M.C.; Almli, L.M.; Ratanatharathorn, A.; Uddin, M.; Galea, S.; Wildman, D.E.; Aiello, A.E.; Bradley, B.; Ressler, K.; et al. Frequency of alcohol consumption in humans; the role of metabotropic glutamate receptors and downstream signaling pathways. Transl. Psychiatry 2015, 5, e586. [Google Scholar] [CrossRef] [PubMed]
  46. Nixon, K.; McClain, J.A. Adolescence as a critical window for developing an alcohol use disorder: Current findings in neuroscience. Curr. Opin. Psychiatry 2010, 23, 227–232. [Google Scholar] [CrossRef] [PubMed]
  47. Peltier, M.R.; Verplaetse, T.L.; Mineur, Y.S.; Petrakis, I.L.; Cosgrove, K.P.; Picciotto, M.R.; McKee, S.A. Sex differences in stress-related alcohol use. Neurobiol. Stress 2019, 10, 100149. [Google Scholar] [CrossRef] [PubMed]
  48. Smart, K.; Cox, S.M.L.; Scala, S.G.; Tippler, M.; Jaworska, N.; Boivin, M.; Séguin, J.R.; Benkelfat, C.; Leyton, M. Sex differences in [11C]ABP688 binding: A positron emission tomography study of mGlu5 receptors. Eur. J. Nucl. Med. Mol. Imaging 2019, 46, 1179–1183. [Google Scholar] [CrossRef]
  49. Zerbib, F.; Bruley des Varannes, S.; Roman, S.; Tutuian, R.; Galmiche, J.-P.; Mion, F.; Tack, J.; Malfertheiner, P.; Keywood, C. Randomised clinical trial: Effects of monotherapy with ADX10059, a mGluR5 inhibitor, on symptoms and reflux events in patients with gastro-oesophageal reflux disease. Aliment. Pharmacol. Ther. 2011, 33, 911–921. [Google Scholar] [CrossRef]
  50. Cleva, R.M.; Olive, M.F. Positive allosteric modulators of type 5 metabotropic glutamate receptors (mGluR5) and their therapeutic potential for the treatment of CNS disorders. Molecules 2011, 16, 2097–2106. [Google Scholar] [CrossRef]
  51. Valyear, M.D.; Villaruel, F.R.; Chaudhri, N. Alcohol-seeking and relapse: A focus on incentive salience and contextual conditioning. Behav. Process. 2017, 141, 26–32. [Google Scholar] [CrossRef] [PubMed]
  52. Martin-Fardon, R.; Weiss, F. Modeling Relapse in Animals. In Behavioral Neurobiology of Alcohol Addiction; Springer: Berlin/Heidelberg, Germany, 2012; pp. 403–432. [Google Scholar]
  53. Gass, J.T.; Trantham-Davidson, H.; Kassab, A.S.; Glen, W.B.; Olive, M.F.; Chandler, L.J.; Chandler, L.J. Enhancement of extinction learning attenuates ethanol-seeking behavior and alters plasticity in the prefrontal cortex. J. Neurosci. 2014, 34, 7562–7574. [Google Scholar] [CrossRef] [PubMed]
  54. Sinclair, C.M.; Cleva, R.M.; Hood, L.E.; Olive, M.F.; Gass, J.T. mGluR5 receptors in the basolateral amygdala and nucleus accumbens regulate cue-induced reinstatement of ethanol-seeking behavior. Pharmacol. Biochem. Behav. 2012, 101, 329–335. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Schroeder, J.P.; Spanos, M.; Stevenson, J.R.; Besheer, J.; Salling, M.; Hodge, C.W. Cue-induced reinstatement of alcohol-seeking behavior is associated with increased ERK1/2 phosphorylation in specific limbic brain regions: Blockade by the mGluR5 antagonist MPEP. Neuropharmacology 2008, 55, 546–554. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Lee, J.-Y.; Choe, E.S.; Yang, C.H.; Choi, K.H.; Cheong, J.H.; Jang, C.-G.; Seo, J.-W.; Yoon, S.S. The mGluR5 antagonist MPEP suppresses the expression and reinstatement, but not the acquisition, of the ethanol-conditioned place preference in mice. Pharmacol. Biochem. Behav. 2016, 140, 33–38. [Google Scholar] [CrossRef] [PubMed]
  57. Adams, C.L.; Cowen, M.S.; Short, J.L.; Lawrence, A.J. Combined antagonism of glutamate mGlu5 and adenosine A2A receptors interact to regulate alcohol-seeking in rats. Int. J. Neuropsychopharmacol. 2008, 11, 229–241. [Google Scholar] [CrossRef] [PubMed]
  58. Kotlinska, J.H.; Bochenski, M.; Danysz, W. The role of group I mGlu receptors in the expression of ethanol-induced conditioned place preference and ethanol withdrawal seizures in rats. Eur. J. Pharmacol. 2011, 670, 154–161. [Google Scholar] [CrossRef] [PubMed]
  59. McGeehan, A.J.; Olive, M.F. The mGluR5 antagonist MPEP reduces the conditioned rewarding effects of cocaine but not other drugs of abuse. Synapse 2003, 47, 240–242. [Google Scholar] [CrossRef] [PubMed]
  60. Simonyi, A.; Schachtman, T.R.; Christoffersen, G.R.J. Metabotropic glutamate receptor subtype 5 antagonism in learning and memory. Eur. J. Pharmacol. 2010, 639, 17–25. [Google Scholar] [CrossRef] [Green Version]
  61. Slattery, D.A.; Neumann, I.D.; Flor, P.J.; Zoicas, I. Pharmacological modulation of metabotropic glutamate receptor subtype 5 and 7 impairs extinction of social fear in a time-point-dependent manner. Behav. Brain Res. 2017, 328, 57–61. [Google Scholar] [CrossRef]
  62. Sethna, F.; Wang, H. Pharmacological enhancement of mGluR5 facilitates contextual fear memory extinction. Learn. Mem. 2014, 21, 647–650. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Sethna, F.; Wang, H. Acute inhibition of mGluR5 disrupts behavioral flexibility. Neurobiol. Learn. Mem. 2016, 130, 1–6. [Google Scholar] [CrossRef] [PubMed]
  64. Fontanez-Nuin, D.E.; Santini, E.; Quirk, G.J.; Porter, J.T. Memory for fear extinction requires mGluR5-mediated activation of infralimbic neurons. Cereb. Cortex 2011, 21, 727–735. [Google Scholar] [CrossRef] [PubMed]
  65. Martínez-Rivera, A.; Rodríguez-Borrero, E.; Matías-Alemán, M.; Montalvo-Acevedo, A.; Guerrero-Figuereo, K.; Febo-Rodríguez, L.J.; Morales-Rivera, A.; Maldonado-Vlaar, C.S. Metabotropic glutamate receptor 5 within nucleus accumbens shell modulates environment-elicited cocaine conditioning expression. Pharmacol. Biochem. Behav. 2013, 110, 154–160. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Herrold, A.A.; Voigt, R.M.; Napier, T.C. mGluR5 is necessary for maintenance of methamphetamine-induced associative learning. Eur. Neuropsychopharmacol. 2013, 23, 691–696. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Marszalek-Grabska, M.; Gibula-Bruzda, E.; Bodzon-Kulakowska, A.; Suder, P.; Gawel, K.; Talarek, S.; Listos, J.; Kedzierska, E.; Danysz, W.; Kotlinska, J.H. ADX-47273, a mGlu5 receptor positive allosteric modulator, attenuates deficits in cognitive flexibility induced by withdrawal from ‘binge-like’ ethanol exposure in rats. Behav. Brain Res. 2018, 338, 9–16. [Google Scholar] [CrossRef] [PubMed]
  68. Bertholomey, M.L.; Nagarajan, V.; Torregrossa, M.M. Sex differences in reinstatement of alcohol seeking in response to cues and yohimbine in rats with and without a history of adolescent corticosterone exposure. Psychopharmacology 2016, 233, 2277–2287. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Coffey, S.F.; Saladin, M.E.; Drobes, D.J.; Brady, K.T.; Dansky, B.S.; Kilpatrick, D.G. Trauma and substance cue reactivity in individuals with comorbid posttraumatic stress disorder and cocaine or alcohol dependence. Drug Alcohol Depend. 2002, 65, 115–127. [Google Scholar] [CrossRef]
  70. Nesic, J.; Duka, T. Gender specific effects of a mild stressor on alcohol cue reactivity in heavy social drinkers. Pharmacol. Biochem. Behav. 2006, 83, 239–248. [Google Scholar] [CrossRef]
  71. Thomas, S.E.; Randall, P.K.; Brady, K.; See, R.E.; Drobes, D.J. An acute psychosocial stressor does not potentiate alcohol cue reactivity in non-treatment-seeking alcoholics. Alcohol. Clin. Exp. Res. 2011, 35, 464–473. [Google Scholar] [CrossRef]
  72. Torres, O.V.; Walker, E.M.; Beas, B.S.; O’Dell, L.E. Female rats display enhanced rewarding effects of ethanol that are hormone dependent. Alcohol. Clin. Exp. Res. 2014, 38, 108–115. [Google Scholar] [CrossRef] [PubMed]
  73. Melón, L.C.; Nolan, Z.T.; Colar, D.; Moore, E.M.; Boehm, S.L. II Activation of extrasynaptic δ-GABAA receptors globally or within the posterior-VTA has estrous-dependent effects on consumption of alcohol and estrous-independent effects on locomotion. Horm. Behav. 2017, 95, 65–75. [Google Scholar] [CrossRef] [PubMed]
  74. O’Leary, O.F.; Cryan, J.F. Towards translational rodent models of depression. Cell Tissue Res. 2013, 354, 141–153. [Google Scholar] [CrossRef] [PubMed]
  75. Lee, K.M.; Coelho, M.A.; Class, M.A.; Szumlinski, K.K. mGlu5-dependent modulation of anxiety during early withdrawal from binge-drinking in adult and adolescent male mice. Drug Alcohol Depend. 2018, 184, 1–11. [Google Scholar] [CrossRef] [PubMed]
  76. Lee, K.M.; Coelho, M.A.; Class, M.A.; Sern, K.R.; Bocz, M.D.; Szumlinski, K.K. mGlu5 Receptor Blockade Within the Nucleus Accumbens Shell Reduces Behavioral Indices of Alcohol Withdrawal-Induced Anxiety in Mice. Front. Pharmacol. 2018, 9, 1306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Lee, K.M.; Coelho, M.A.; Sern, K.R.; Class, M.A.; Bocz, M.D.; Szumlinski, K.K. Anxiolytic Effects of Buspirone and MTEP in the Porsolt Forced Swim Test. Chronic Stress 2017, 1, 2470547017712985. [Google Scholar] [CrossRef]
  78. Hales, C.A.; Stuart, S.A.; Anderson, M.H.; Robinson, E.S.J. Modelling cognitive affective biases in major depressive disorder using rodents. Br. J. Pharmacol. 2014, 171, 4524–4538. [Google Scholar] [CrossRef] [Green Version]
  79. Esterlis, I.; Holmes, S.E.; Sharma, P.; Krystal, J.H.; DeLorenzo, C. Metabotropic Glutamatergic Receptor 5 and Stress Disorders: Knowledge Gained From Receptor Imaging Studies. Biol. Psychiatry 2018, 84, 95–105. [Google Scholar] [CrossRef]
  80. Chandley, M.J.; Szebeni, A.; Szebeni, K.; Crawford, J.D.; Stockmeier, C.A.; Turecki, G.; Kostrzewa, R.M.; Ordway, G.A. Elevated gene expression of glutamate receptors in noradrenergic neurons from the locus coeruleus in major depression. Int. J. Neuropsychopharmacol. 2014, 17, 1569–1578. [Google Scholar] [CrossRef] [Green Version]
  81. Esterlis, I.; DellaGioia, N.; Pietrzak, R.H.; Matuskey, D.; Nabulsi, N.; Abdallah, C.G.; Yang, J.; Pittenger, C.; Sanacora, G.; Krystal, J.H.; et al. Ketamine-induced reduction in mGluR5 availability is associated with an antidepressant response: An [11C]ABP688 and PET imaging study in depression. Mol. Psychiatry 2017, 23, 824–832. [Google Scholar] [CrossRef]
  82. Deschwanden, A.; Karolewicz, B.; Feyissa, A.M.; Treyer, V.; Ametamey, S.M.; Johayem, A.; Burger, C.; Auberson, Y.P.; Sovago, J.; Stockmeier, C.A.; et al. Reduced Metabotropic Glutamate Receptor 5 Density in Major Depression Determined by [11C]ABP688 PET and Postmortem Study. Am. J. Psychiatry 2011, 168, 727–734. [Google Scholar] [CrossRef] [PubMed]
  83. Abdallah, C.G.; Hannestad, J.; Mason, G.F.; Holmes, S.E.; DellaGioia, N.; Sanacora, G.; Jiang, L.; Matuskey, D.; Satodiya, R.; Gasparini, F.; et al. Metabotropic Glutamate Receptor 5 and Glutamate Involvement in Major Depressive Disorder: A Multimodal Imaging Study. Biol. Psychiatry Cogn. Neurosci. Neuroimaging 2017, 2, 449–456. [Google Scholar] [CrossRef] [PubMed]
  84. Matosin, N.; Fernandez-Enright, F.; Frank, E.; Deng, C.; Wong, J.; Huang, X.-F.; Newell, K. Metabotropic glutamate receptor mGluR2/3 and mGluR5 binding in the anterior cingulate cortex in psychotic and nonpsychotic depression, bipolar disorder and schizophrenia: Implications for novel mGluR-based therapeutics. J. Psychiatry Neurosci. 2014, 39, 407–416. [Google Scholar] [CrossRef] [PubMed]
  85. Fatemi, S.H.; Folsom, T.D.; Rooney, R.J.; Thuras, P.D. mRNA and protein expression for novel GABAA receptors θ and ρ2 are altered in schizophrenia and mood disorders; relevance to FMRP-mGluR5 signaling pathway. Transl. Psychiatry 2013, 3, e271. [Google Scholar] [CrossRef] [PubMed]
  86. DeLorenzo, C.; Sovago, J.; Gardus, J.; Xu, J.; Yang, J.; Behrje, R.; Kumar, J.S.D.; Devanand, D.P.; Pelton, G.H.; Mathis, C.A.; et al. Characterization of brain mGluR5 binding in a pilot study of late-life major depressive disorder using positron emission tomography and [11C]ABP688. Transl. Psychiatry 2015, 5, 1–7. [Google Scholar] [CrossRef] [PubMed]
  87. AstraZeneca 6-week Study Treatment to Evaluate the Safety and Effectiveness of AZD2066 in Patients with Major Depressive Disorder. Available online: https://clinicaltrials.gov/ct2/show/NCT01145755 (accessed on 22 July 2019).
  88. Quiroz, J.A.; Tamburri, P.; Deptula, D.; Banken, L.; Beyer, U.; Rabbia, M.; Parkar, N.; Fontoura, P.; Santarelli, L. Efficacy and Safety of Basimglurant as Adjunctive Therapy for Major Depression. JAMA Psychiatry 2016, 73, 675–684. [Google Scholar] [CrossRef] [PubMed]
  89. Mohammad, F.; Ho, J.; Woo, J.H.; Lim, C.L.; Poon, D.J.J.; Lamba, B.; Claridge-Chang, A. Concordance and incongruence in preclinical anxiety models: Systematic review and meta-analyses. Neurosci. Biobehav. Rev. 2016, 68, 504–529. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Thomas, A.; Burant, A.; Bui, N.; Graham, D.; Yuva-Paylor, L.A.; Paylor, R. Marble burying reflects a repetitive and perseverative behavior more than novelty-induced anxiety. Psychopharmacology 2009, 204, 361–373. [Google Scholar] [CrossRef] [Green Version]
  91. Albelda, N.; Joel, D. Animal models of obsessive-compulsive disorder: Exploring pharmacology and neural substrates. Neurosci. Biobehav. Rev. 2012, 36, 47–63. [Google Scholar] [CrossRef]
  92. Kotlinska, J.; Bochenski, M. The influence of various glutamate receptors antagonists on anxiety-like effect of ethanol withdrawal in a plus-maze test in rats. Eur. J. Pharmacol. 2008, 598, 57–63. [Google Scholar] [CrossRef]
  93. Kumar, J.; Hapidin, H.; Bee, Y.-T.G.; Ismail, Z. Effects of the mGluR5 antagonist MPEP on ethanol withdrawal induced anxiety-like syndrome in rats. Behav. Brain Funct. 2013, 9, 43. [Google Scholar] [CrossRef] [PubMed]
  94. Griebel, G.; Holmes, A. 50 years of hurdles and hope in anxiolytic drug discovery. Nat. Rev. Drug Discov. 2013, 12, 667–687. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Riaza Bermudo-Soriano, C.; Perez-Rodriguez, M.M.; Vaquero-Lorenzo, C.; Baca-Garcia, E. New perspectives in glutamate and anxiety. Pharmacol. Biochem. Behav. 2012, 100, 752–774. [Google Scholar] [CrossRef] [PubMed]
  96. Lee, K.M.; Coehlo, M.A.; Solton, N.R.; Szumlinski, K.K. Negative Affect and Excessive Alcohol Intake Incubate during Protracted Withdrawal from Binge-Drinking in Adolescent, But Not Adult, Mice. Front. Psychol. 2017, 8, 1128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Loxton, D.; Canales, J.J. Long-term cognitive, emotional and neurogenic alterations induced by alcohol and methamphetamine exposure in adolescent rats. Prog. Neuropsychopharmacol. Biol. Psychiatry 2017, 74, 1–8. [Google Scholar] [CrossRef] [PubMed]
  98. Rico-Barrio, I.; Peñasco, S.; Puente, N.; Ramos, A.; Fontaine, C.J.; Reguero, L.; Giordano, M.E.; Buceta, I.; Terradillos, I.; Lekunberri, L.; et al. Cognitive and neurobehavioral benefits of an enriched environment on young adult mice after chronic ethanol consumption during adolescence. Addict. Biol. 2018, 14. [Google Scholar] [CrossRef] [PubMed]
  99. Szumlinski, K.K.; Coelho, M.A.; Lee, K.M.; Tran, T.; Sern, K.R.; Bernal, A.; Kippin, T.E. DID it or DIDn’t it? Exploration of a failure to replicate binge-like alcohol-drinking in C57BL/6J mice. Pharmacol. Biochem. Behav. 2019, 178, 3–18. [Google Scholar] [CrossRef]
  100. Van Skike, C.E.; Diaz-Granados, J.L.; Matthews, D.B. Chronic Intermittent Ethanol Exposure Produces Persistent Anxiety in Adolescent and Adult Rats. Alcohol. Clin. Exp. Res. 2015, 39, 262–271. [Google Scholar] [CrossRef] [Green Version]
  101. Lee, K.M.; Coelho, M.A.; Sern, K.R.; Szumlinski, K.K. Homer2 within the central nucleus of the amygdala modulates withdrawal-induced anxiety in a mouse model of binge-drinking. Neuropharmacology 2018, 128, 448–459. [Google Scholar] [CrossRef]
  102. Van Waes, V.; Darnaudéry, M.; Marrocco, J.; Gruber, S.H.; Talavera, E.; Mairesse, J.; Van Camp, G.; Casolla, B.; Nicoletti, F.; Mathé, A.A.; et al. Impact of early life stress on alcohol consumption and on the short- and long-term responses to alcohol in adolescent female rats. Behav. Brain Res. 2011, 221, 43–49. [Google Scholar] [CrossRef]
  103. Grueter, B.A.; Gosnell, H.B.; Olsen, C.M.; Schramm-Sapyta, N.L.; Nekrasova, T.; Landreth, G.E.; Winder, D.G. Extracellular-Signal Regulated Kinase 1-Dependent Metabotropic Glutamate Receptor 5-Induced Long-Term Depression in the Bed Nucleus of the Stria Terminalis Is Disrupted by Cocaine Administration. J. Neurosci. 2006, 26, 3210–3219. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Grueter, B.A.; McElligott, Z.A.; Robison, A.J.; Mathews, G.C.; Winder, D.G. In Vivo Metabotropic Glutamate Receptor 5 (mGluR5) Antagonism Prevents Cocaine-Induced Disruption of Postsynaptically Maintained mGluR5-Dependent Long-Term Depression. J. Neurosci. 2008, 28, 9261–9270. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Wolf, M.E.; Tseng, K.Y. Calcium-permeable AMPA receptors in the VTA and nucleus accumbens after cocaine exposure: When, how, and why? Front. Mol. Neurosci. 2012, 5, 72. [Google Scholar] [CrossRef] [PubMed]
  106. Loweth, J.A.; Tseng, K.Y.; Wolf, M.E. Using metabotropic glutamate receptors to modulate cocaine’s synaptic and behavioral effects: mGluR1 finds a niche. Curr. Opin. Neurobiol. 2013, 23, 500–506. [Google Scholar] [CrossRef] [PubMed]
  107. Loweth, J.A.; Tseng, K.Y.; Wolf, M.E. Adaptations in AMPA receptor transmission in the nucleus accumbens contributing to incubation of cocaine craving. Neuropharmacology 2014, 76, 287–300. [Google Scholar] [CrossRef] [PubMed]
  108. Ma, Y.-Y.; Lee, B.R.; Wang, X.; Guo, C.; Liu, L.; Cui, R.; Lan, Y.; Balcita-Pedicino, J.J.; Wolf, M.E.; Sesack, S.R.; et al. Bidirectional Modulation of Incubation of Cocaine Craving by Silent Synapse-Based Remodeling of Prefrontal Cortex to Accumbens Projections. Neuron 2014, 83, 1453–1467. [Google Scholar] [CrossRef] [Green Version]
  109. Szumlinski, K.K.; Lominac, K.D.; Oleson, E.B.; Walker, J.K.; Mason, A.; Dehoff, M.H.; Klugmann, M.; Klugman, M.; Cagle, S.; Welt, K.; et al. Homer2 Is Necessary for EtOH-Induced Neuroplasticity. J. Neurosci. 2005, 25, 7054–7061. [Google Scholar] [CrossRef] [Green Version]
  110. Szumlinski, K.K.; Ary, A.W.; Lominac, K.D.; Klugmann, M.; Kippin, T.E. Accumbens Homer2 overexpression facilitates alcohol-induced neuroplasticity in C57BL/6J mice. Neuropsychopharmacology 2008, 33, 1365–1378. [Google Scholar] [CrossRef]
  111. Cozzoli, D.K.; Courson, J.; Caruana, A.L.; Miller, B.W.; Greentree, D.I.; Thomspon, A.B.; Wroten, M.G.; Zhang, P.-W.; Xiao, B.; Hu, J.-H.; et al. Nucleus Accumbens mGluR5-Associated Signaling Regulates Binge Alcohol Drinking Under Drinking-in-the-Dark Procedures. Alcohol. Clin. Exp. Res. 2012, 36, 1623–1633. [Google Scholar] [CrossRef]
  112. Cozzoli, D.K.; Kaufman, M.N.; Nipper, M.A.; Hashimoto, J.G.; Wiren, K.M.; Finn, D.A. Functional regulation of PI3K-associated signaling in the accumbens by binge alcohol drinking in male but not female mice. Neuropharmacology 2016, 105, 164–174. [Google Scholar] [CrossRef] [Green Version]
  113. Lum, E.N.; Campbell, R.R.; Rostock, C.; Szumlinski, K.K. mGluR1 within the nucleus accumbens regulates alcohol intake in mice under limited-access conditions. Neuropharmacology 2014, 79, 679–687. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Martinez, L.A.; Peterson, B.M.; Meisel, R.L.; Mermelstein, P.G. Estradiol facilitation of cocaine-induced locomotor sensitization in female rats requires activation of mGluR5. Behav. Brain Res. 2014, 271, 39–42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Martinez, L.A.; Gross, K.S.; Himmler, B.T.; Emmitt, N.L.; Peterson, B.M.; Zlebnik, N.E.; Foster Olive, M.; Carroll, M.E.; Meisel, R.L.; Mermelstein, P.G. Estradiol Facilitation of Cocaine Self-Administration in Female Rats Requires Activation of mGluR5. eNeuro 2016, 3. [Google Scholar] [CrossRef] [PubMed]
  116. Tonn Eisinger, K.R.; Gross, K.S.; Head, B.P.; Mermelstein, P.G. Interactions between estrogen receptors and metabotropic glutamate receptors and their impact on drug addiction in females. Horm. Behav. 2018, 104, 130–137. [Google Scholar] [CrossRef] [PubMed]
Table 1. Details from studies on the effects of mGlu5 receptor modulation on ethanol intake in continuous access, limited access, and operant ethanol drinking paradigms.
Table 1. Details from studies on the effects of mGlu5 receptor modulation on ethanol intake in continuous access, limited access, and operant ethanol drinking paradigms.
Continuous Access
ManipulationAverage Reported Ethanol IntakeTreatment DetailsSpecies/Strain/SexHousingEffectDoseReference
GRM5 mutationTS/TS: greater than 6.0 g/kg Male & female GRM5TS/TS, TS/AA, AA/AA miceGroupedIncreasedAA/AA[19]
MTEPUp to 20 g/kgRepeated systemic prior to accessFemale B6 miceIndividualIncreased & Decreased20 mg/kg[31]
mGlu5 receptor deficiencyWild type: greater than 9.0 g/kg Male Grm5tm1Rod mice Decreasedn/a[17]
mGlu5 receptor knockoutWild type: up to 3.0 g/kg Female mGlu5−/− mice Decreased, no changen/a[32]
MTEPUp to 5 g/kgRepeated systemicMale FH rats Decreased2 mg/kg[27]
MTEPUp to 15 g/kgRepeated systemic prior to accessMale B6 miceIndividualDecreased20 mg/kg[31]
MPEP0.53 ± 0.05 g/kgRepeated systemicMale Wistar ratsIndividualDecreased3, 10 mg/kg[30]
MPEPGreater than 5.0 g/kgRepeated systemicMeyers ratsIndividualDecreased1, 3 mg/kg[33]
MPEP17.9 ± 8.2 g/kgRepeated systemicMale B6 miceIndividualDecreased10 mg/kg[25]
mGlu5 receptor knockdown on D1 neuronsUp to 6 g/kg Male mGlu5KD−D1 miceIndividualNo changen/a[16]
mGlu5 receptor knockoutWild type: up to 2.0 g/kg Male mGlu5−/− mice No changen/a[32]
Impaired mGlu5/Homer interactionWild type: 10.84 ± 2.26 g/kg Male mGlu5-F1128R miceGroupedNo changen/a[18]
Limited Access
ManipulationAverage Reported Ethanol IntakeTreatment DetailsSpecies/Strain/SexHousingEffectDoseReference
mGlu5 receptor knockoutWild type: greater than 2.0 g/kg Female mGlu5−/− mice Decreasedn/a[32]
MTEPUp to 3 g/kgRepeated systemicFemale B6 miceIndividualDecreased20 mg/kg[31]
MTEPUp to 3.5 g/kgRepeated systemicMale B6 miceIndividualDecreased10, 20 mg/kg[31]
MTEPUp to 4.5 g/kgAcute intra-CeAMale B6 miceIndividualDecreased3 µg/side[34]
MPEPUp to 1.5 g/kgAcute intra-NAcMale B6 miceIndividualDecreased0.1, 0.3, 1 µg/side[18]
mGlu5 receptor knockoutWild type: up to 1.5 g/kg Female mGlu5−/− mice No changen/a[32]
MTEPUp to 2.0 g/kgAcute intra-adBNSTMale & female GRM5TS/TS, TS/AA, AA/AA miceIndividualNo change30 µg/side[19]
MPEPGreater than 0.75 g/kgAcute intra-NAcMale mGlu5-F1128R miceIndividualNo change1 µg/side[18]
Operant Responding
ManipulationAverage Reported Ethanol IntakeTreatment DetailsSpecies/Strain/SexHousingEffectDoseReference
GRM5 mutationTS/TS: up to 1.5 g/kg Male & female GRM5TS/TS, TS/AA, AA/AA miceGroupedIncreasedAA/AA[19]
mGlu5 receptor knockdown on D1 neuronsWild type: up to 3000 licks Female mGlu5KD−D1 miceGroupedDecreasedn/a[15]
MTEPUp to 80 responsesAcute systemicMale FH rats Decreased2 mg/kg[27]
MTEPGreater than 100 responsesAcute systemicMale iP rats Decreased1, 2 mg/kg[27]
MTEPGreater than 100 responsesAcute systemicMale B6 miceGroupedDecreased20, 40 mg/kg[28]
MTEPUp to 20 responsesAcute intra-NAc shellMale Wistar rats Decreased1.5 µg/side[35]
MTEPNon-dependent: up to 30 responses
Dependent: up to 40 responses
Acute systemicMale Wistar ratsGroupedDecreased1, 3 mg/kg[36]
MPEPUp to 80 responsesAcute systemicMale iP ratsIndividualDecreased3, 10 mg/kg[24]
MPEPGreater than 8.0 g/kgAcute systemicMale B6 miceIndividualDecreased3, 10 mg/kg[25]
MPEPUp to 5 g/kgAcute systemicMale B6 mice Decreased3, 10 mg/kg[26]
MPEPGreater than 0.6 g/kgAcute systemicMale iP ratsPairDecreased3, 10 mg/kg[29]
MPEP0.96 ± 0.22 g/kgAcute intra-NAc medial coreMale iP ratsIndividualDecreased10 µg/side[37]
MTEPUp to 15 responsesAcute intra-NAc coreMale Wistar rats No change1.5 µg/side[35]
MTEP0.60 ± 0.1 g/kgAcute systemiciP ratsPairNo change2.5 mg/kg[23]
MPEP1.15 ± 0.18 g/kgAcute intra-dorsomedial caudateMale iP ratsIndividualNo change1, 3, 10 µg/side[37]
MPEP1.02 ± 0.08 g/kgAcute intra-medial PFCMale iP ratsIndividualNo change1, 3, 10, 30 µg/side[37]
wild-type (GRM5TS/TS), heterozygous mutant (GRM5TS/AA), homozygous mutant (GRM5AA/AA), C57Bl/6J (B6), Fawn Hooded (FH), central amygdala (CeA), nucleus accumbens (NAc), anterior dorsal bed nucleus of the stria terminalis (adBNST), inbred alcohol-preferring (iP), prefrontal cortex (PFC), 2-Methyl-6-(phenylethynyl)pyridine (MPEP) and 3-((2-Methyl-1,3-thiazol-4-yl)ethynyl)pyridine (MTEP).
Table 2. Details from studies observing the effect of mGlu5 receptor modulation on cue-induced reinstatement to seek ethanol and ethanol-conditioned place preference.
Table 2. Details from studies observing the effect of mGlu5 receptor modulation on cue-induced reinstatement to seek ethanol and ethanol-conditioned place preference.
Ethanol Cue-Induced Reinstatement
ManipulationAverage Reported Ethanol IntakeTreatment DetailsSpecies/Strain/SexHousingEffectDoseReference
CDPPBUp to 80 responsesRepeated systemic, during extinctionMale Wistar ratsIndividualDecreased20 mg/kg[53]
MTEPGreater than 60 responsesAcute intra-BLA, prior to reinstatement testMale Wistar ratsIndividualDecreased3.0 µg/µl[54]
MTEPGreater than 40 responsesAcute intra-NAc core, prior to reinstatement testMale Wistar ratsIndividualDecreased3.0 µg/µl[54]
MPEPUp to 60 responsesAcute systemic, prior to reinstatement testMale iP ratsPairDecreased1, 10 mg/kg[55]
MPEP0.53 ± 0.05 g/kgAcute systemic, prior to reinstatement testMale Long Evans ratsPairDecreased3, 10 mg/kg[30]
MPEP2.0 g/kgAcute systemic, prior to reinstatement testMale B6 miceGroupedDecreased20 mg/kg[56]
MTEP0.54 ± 0.04 g/kgAcute systemic, prior to reinstatement testiP ratsPairNo change2.5 mg/kg[57]
MTEP0.60 ± 0.1 g/kgAcute systemic, prior to reinstatement testiP ratsPairNo change2.5 mg/kg[23]
Ethanol Conditioned Place Preference
ManipulationEthanol DoseTreatment DetailsSpecies/Strain/SexHousingEffectDoseReference
GRM5 mutation1.0–3.0 g/kg Male & female GRM5TS/TS, TS/AA, AA/AA miceGroupedIncreased
Decreased
TS/TS
AA/AA
[19]
mGlu5 receptor deficiency1.0 g/kg Male Grm5tm1Rod mice Decreasedn/a[17]
MTEP0.5 g/kgAcute systemic, prior to testMale Wistar ratsGroupedDecreased2.5, 5 mg/kg[58]
MPEP2.0 g/kgAcute systemic, prior to testMale B6 miceGroupedDecreased20 mg/kg[56]
MPEP2.0 g/kgAcute systemic, prior to testMale B6 miceIndividualDecreased10 mg/kg[25]
mGlu5 receptor knockdown on D1 neurons1.5 g/kg Male & female mGlu5KD−D1 miceGroupedNo changen/a[15]
MPEP2.0 g/kgRepeated systemic, during acquisitionMale B6 miceGroupedNo change5, 10, 20 mg/kg[56]
MPEP2.0 g/kgRepeated systemic, during acquisitionMale D2 miceGroupedNo change1, 5, 20 mg/kg[59]
Basolateral amygdala (BLA), nucleus accumbens (NAc), inbred preferring rat (iP), C57Bl/6J (B6), DBA/2J (D2), 3-Cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl)benzamide (CDPPB).
Table 3. Details of the effects of mGlu5 receptor modulation on behavioral despair in the forced swim task.
Table 3. Details of the effects of mGlu5 receptor modulation on behavioral despair in the forced swim task.
ManipulationAverage Reported Ethanol Intake Treatment DetailsSpecies/Strain/SexHousingAlcohol × Drug EffectDoseReference
MTEP4.00 ± 0.05 g/kgAcute systemicAdult male B6 miceGroupedRescued30 mg/kg[77]
MTEPGreater than 4.0 g/kgAcute systemicAdult male B6 miceGroupedRescued30 mg/kg[75]
CDPPBGreater than 4.0 g/kgAcute systemicAdult male B6 miceGroupedExacerbated30 mg/kg[75]
MTEPGreater than 5.0 g/kgAcute systemicAdolescent male B6 miceGroupedNo change30 mg/kg[75]
MTEPUp to 7.0 g/kgAcute intra-NAc shellAdolescent male B6 miceGroupedNo change1, 10 µg/side[76]
MTEPUp to 5.0 g/kgAcute intra-NAc shellAdult male B6 miceGroupedNo change1, 10 µg/side[76]
CDPPBGreater than 5.0 g/kgAcute systemicAdolescent male B6 miceGroupedNo change30 mg/kg[75]
C57Bl/6J (B6), nucleus accumbens (NAc).
Table 4. Details from studies assessing anxiety-like activity following mGlu5 modulation in the elevated plus maze (EPM), light/dark box (LD), open field (OF), and marble burying (MB) tasks.
Table 4. Details from studies assessing anxiety-like activity following mGlu5 modulation in the elevated plus maze (EPM), light/dark box (LD), open field (OF), and marble burying (MB) tasks.
ManipulationTaskAverage Reported Ethanol IntakeTreatment DetailsSpecies/Strain/SexHousingAlcohol × Drug EffectDoseReference
MTEPEPMUp to 2 g/kgAcute systemicAdult male Wistar ratsGroupedRescued2.5, 5 mg/kg[92]
MPEPEPMGreater than 10.0 g/kgAcute systemicMale Wistar ratsIndividualRescued2.5, 5, 10, 20, 30 mg/kg[93]
MTEPLDGreater than 4.0 g/kgAcute systemicAdult male B6 miceGroupedRescued30 mg/kg[75]
MTEPLDUp to 5.0 g/kgAcute intra-NAc shellAdult male B6 miceGroupedRescued1 µg/side[76]
CDPPBLDGreater than 4.0 g/kgAcute systemicAdult male B6 miceGroupedExacerbated30 mg/kg[75]
MTEPLDGreater than 5.0 g/kgAcute systemicAdolescent male B6 miceGroupedNo change30 mg/kg[75]
CDPPBLDGreater than 5.0 g/kgAcute systemicAdolescent male B6 miceGroupedNo change30 mg/kg[75]
MTEPLDUp to 7.0 g/kgAcute intra-NAc shellAdolescent male B6 miceGroupedNo change1, 10 µg/side[76]
MPEPOFGreater than 10.0 g/kgAcute systemicMale Wistar ratsIndividualRescued2.5, 5, 10 mg/kg[93]
MTEPMBGreater than 4.0 g/kgAcute systemicAdult male B6 miceGroupedRescued30 mg/kg[75]
MTEPMBUp to 5.0 g/kgAcute intra-NAc shellAdult male B6 miceGroupedRescued1 µg/side[76]
MTEPMBUp to 7.0 g/kgAcute intra-NAc shellAdolescent male B6 miceGroupedRescued10 µg/side[76]
MTEPMBGreater than 5.0 g/kgAcute systemicAdolescent male B6 miceGroupedDecreased30 mg/kg[75]
CDPPBMBGreater than 5.0 g/kgAcute systemicAdolescent male B6 miceGroupedIncreased 30 mg/kg[75]
CDPPBMBGreater than 4.0 g/kgAcute systemicAdult male B6 miceGroupedNo change30 mg/kg[75]
Elevated plus maze (EPM), light/dark box (LD), C57Bl/6J (B6), nucleus accumbens (NAc), open field (OF), marble burying (MB).

Share and Cite

MDPI and ACS Style

Kasten, C.R.; Holmgren, E.B.; Wills, T.A. Metabotropic Glutamate Receptor Subtype 5 in Alcohol-Induced Negative Affect. Brain Sci. 2019, 9, 183. https://doi.org/10.3390/brainsci9080183

AMA Style

Kasten CR, Holmgren EB, Wills TA. Metabotropic Glutamate Receptor Subtype 5 in Alcohol-Induced Negative Affect. Brain Sciences. 2019; 9(8):183. https://doi.org/10.3390/brainsci9080183

Chicago/Turabian Style

Kasten, Chelsea R., Eleanor B. Holmgren, and Tiffany A. Wills. 2019. "Metabotropic Glutamate Receptor Subtype 5 in Alcohol-Induced Negative Affect" Brain Sciences 9, no. 8: 183. https://doi.org/10.3390/brainsci9080183

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop