Abstract
Striking sex differences exist in presentation and incidence of several psychiatric disorders. For example, major depressive disorder is more prevalent in women than men, and women who develop alcohol use disorder progress through drinking milestones more rapidly than men. With regards to psychiatric treatment responses, women respond more favorably to selective serotonin reuptake inhibitors than men, whereas men have better outcomes when prescribed tricyclic antidepressants. Despite such well-documented biases in incidence, presentation, and treatment response, sex as a biological variable has long been neglected in preclinical and clinical research. An emerging family of druggable targets for psychiatric diseases, metabotropic glutamate (mGlu) receptors are G-protein coupled receptors broadly distributed throughout the central nervous system. mGlu receptors confer diverse neuromodulatory actions of glutamate at the levels of synaptic plasticity, neuronal excitability, and gene transcription. In this chapter, we summarize the current preclinical and clinical evidence for sex differences in mGlu receptor function. We first highlight basal sex differences in mGlu receptor expression and function and proceed to describe how gonadal hormones, notably estradiol, regulate mGlu receptor signaling. We then describe sex-specific mechanisms by which mGlu receptors differentially modulate synaptic plasticity and behavior in basal states and models relevant for disease. Finally, we discuss human research findings and highlight areas in need of further research. Taken together, this review emphasizes how mGlu receptor function and expression can differ across sex. Gaining a more complete understanding of how sex differences in mGlu receptor function contribute to psychiatric diseases will be critical in the development of novel therapeutics that are effective in all individuals.
1. Introduction
Sex differences in the incidence, presentation, and treatment responses of several psychiatric and substance use disorders are well-known. Major depressive disorder (MDD), for instance, exhibits sex differences across several levels. Women are approximately twice as likely to have MDD than men, exhibit more severe symptoms, and often experience different symptomatology (Angst & Dobler-Mikola, 1984; Frank, Carpenter, & Kupfer, 1988; Kessler et al., 2005; Kornstein et al., 2000a; Najt, Fusar-Poli, & Brambilla, 2011; Silverstein, 1999; Young et al., 1990). Additionally, evidence suggests that women and men, on average, have superior treatment responses to different antidepressant medications. For instance, women have statistically superior responses to monoamine oxidase inhibitors and selective serotonin reuptake inhibitors than men; conversely, men respond more favorably to tricyclic antidepressants (Hamilton, Grant, & Jensvold, 1996; Kornstein et al., 2000b; Quitkin et al., 2002). In contrast, many reported sex differences in schizophrenia are weighted toward men. For example, men have an earlier age of onset of schizophrenia, with symptoms emerging in the early twenties in men, while symptom emergence is in the late twenties in women (Angermeyer, Goldstein, & Kuehn, 1989; Goldstein, Tsuang, & Faraone, 1989; Hafner et al., 1989; Hambrecht, Maurer, & Hafner, 1992; Sartorius et al., 1986). Men with schizophrenia also exhibit higher incidence of negative symptoms and poorer course compared to women with schizophrenia (Goldstein et al., 1998; Goldstein & Link, 1988; Huber et al., 1980; Salokangas, 1983). Similarly, sex-dependent vulnerabilities to specific disease symptoms have been observed in alcohol use disorders (AUD) and several concerning findings indicate that women are at greater risks for detrimental outcomes. While AUDs are more prevalent in men overall, women are more sensitive to peripheral diseases and cognitive disturbances that can stem from chronic alcohol consumption (Nixon, Tivis, & Parsons, 1995; Tuyns & Pequignot, 1984; Urbano-Marquez et al., 1995). Women who develop AUD also progress through disease milestones more rapidly than men (Diehl et al., 2007; Lewis, Hoffman, & Nixon, 2014; Randall et al., 1999), and women are disproportionately affected by consequences of acute intoxication (Gross & Billingham, 1998). Clearly, many psychiatric diseases display remarkable sex differences in presentation and etiology. With the goal of delivering optimal treatment outcomes to all, understanding the manifestation of these sex differences should be a top priority of biological psychiatry research.
Despite the well-documented sex differences in the epidemiology of psychiatric disorders, it is only relatively recently that researchers have begun to delve into potential biological underpinnings for these observed sex differences. A major driving force behind these recent studies is the National Institutes of Health mandate that sex as a biological variable (SABV) will be factored into research designs, analyses, and reporting in vertebrate animal and human studies. Prior to the mandate, many researchers argued that the circulating gonadal hormones associated with the estrous cycle would introduce too much variability into experimental measures; the result is that many previous basic and preclinical studies excluded female subjects and cells. Thus, much of what is known about biological mechanisms of psychiatric disorders is based on studies in males. The NIMH mandate to include SABV has resulted in an explosion of research documenting sex differences in cell signaling, synaptic plasticity, emotional behaviors, cognition, and social function. In this chapter, we describe and summarize the known sex differences in the functions of metabotropic glutamate (mGlu) receptors. Consistent with the broad and rich neuroendocrinology literature documenting acute effects of gonadal hormones on brain function, significant evidence suggests that sex hormones regulate a variety of mGlu receptor functions through proximal receptor-receptor interactions. However, much less is known about how other factors, including developmental hormone exposure and sex chromosome complement, might influence sex-specific behavioral and neurobiological outcomes. In addition to synthesizing key themes from the existing literature, we hope this chapter will be useful in identifying and highlighting the important gaps in our understanding of sex differences in mGlu receptor function.
2. Sex differences in mGlu receptor function
Metabotropic glutamate (mGlu) receptors are a class of type C G protein-coupled receptors widely distributed in the nervous system. Activation of mGlu receptors via extracellular binding of glutamate triggers intracellular signaling cascades that induce short- and long-term adaptations in synaptic physiology, neuron excitability, and gene expression (Niswender & Conn, 2010). The eight mGlu receptor subtypes are grouped into three families (Groups 1–3) based on sequence homology, cell signaling, and pharmacology (Conn & Pin, 1997).
2.1. Regional distribution and receptor expression
The Group I mGlu receptors, comprised of subtypes mGlu1 and mGlu5, are important regulators of excitatory glutamate transmission and synaptic plasticity. mGlu1 and mGlu5 receptors are primarily localized to postsynaptic compartments, where they signal through Gq proteins and associated downstream effectors (Hermans & Challiss, 2001; Niswender & Conn, 2010). Our current understanding of the regional distribution and localization of Group 1 receptors in the CNS is largely based on gene-cloning and immunoreactivity studies from preclinical models (Martin et al., 1992; Romano et al., 1995; Van den Pol, 1994; van den Pol, Romano, & Ghosh, 1995). mGlu1 and mGlu5 receptors have widespread, generally overlapping distributions throughout most of the brain, with a key exception being the cerebellum, where high levels of mGlu1 (but not mGlu5) are expressed in mature Purkinje cells (Baude et al., 1993; Casabona et al., 1997; Fazio et al., 2008). An important caveat to this literature is that most studies investigating the regional distribution of Group 1 receptors were restricted to male rodents or not statistically powered to test for interactions across sexes. As such, the potential for intrinsic sex differences in the regional distribution of mGlu1/5 receptors remains largely unexplored.
Although major sex differences in the regional distribution of Group 1 receptors have not been reported (Wickens, Bangasser, & Briand, 2018), preclinical studies investigating the potential sex differences in levels of mGlu receptor protein expression have revealed conflicting results. In a Western blot analysis of mGlu receptor subunit protein expression, adult female rats exhibited higher levels of mGlu5 expression in the prefrontal cortex (PFC) and hippocampus than males, with no sex differences in mGlu1 receptor protein expression in the PFC or hippocampus (Wang et al., 2015). These findings are in direct contrast to results from a recent study published in 2017, in which female rats had significantly lower levels of mGlu1 receptor protein expression in the hippocampus than males (Ning et al., 2018). Both of these studies used Sprague-Dawley rats, suggesting that differences in animal strain do not account for the divergent findings. Interestingly, however, these studies employed antibodies raised against different portions of the intracellular C-terminal tail; therefore, it is possible that changes in the proportion of splice variants or post-translational modifications might account for the discrepancies in the literature. Additional studies examining cell type-specific expression of mGlu1 and mGlu5, as well as the relative expression of common splice variants and modifications, appear duly warranted.
There are limited published accounts describing sex differences in Group 2 and Group 3 mGlu receptor expression and function. As such, little is known about how these receptor subtypes endogenously differ in male and female brains. Higher levels of mGlu2/3 receptor expression (assessed with a non-specific antibody) were found in the hippocampus of wild-type female rats in comparison to males, and no sex differences were detected in PFC or amygdala (Wang et al., 2015). In addition, a study in human temporal cortex found higher expression of mGlu3 receptors in women relative to men (García-Bea et al., 2016). To our knowledge, sex differences in baseline Group 2 receptor expression have not been thoroughly explored in other brain regions. Similarly, to our knowledge, potential sex differences in regional or synaptic localization of Group 3 receptors have not been thoroughly investigated.
2.2. Receptor trafficking
Group 1 receptors are localized to postsynaptic densities and anchored to nearby receptors through interactions with a multitude of scaffolding proteins, most notably including Homer, Shank, and PSD95 (Tu et al., 1999). There are three genes that comprise the Homer family proteins (Homer1–3) (Brakeman et al., 1997; Xiao et al., 1998) each of which has multiple alternative short and long form splice variants involved in the trafficking and signaling of Group 1 receptors (Kammermeier et al., 2000). Long form splice variants of Group 1 Homer proteins (Homer1b and Homer1c) differentially regulate mGlu1/5 receptor trafficking on the post-synaptic membrane. In cultures from male rodents, Homer1b inhibits mGlu5 receptor trafficking to the cell surface (Roche et al., 1999), whereas co-expression of Homer1c increases surface expression of mGlu1 receptors (Ciruela et al., 2000; Xiao, Gustafsson, & Niu, 2006). To our knowledge, these findings have not been replicated in cultures from female animals. Nonetheless, there is little evidence to suggest basal sex differences in Homer protein regulation of Group 1 receptors, and neither Homer1 nor Homer2 knockout mice present with sex-dependent phenotypes in unconditioned behaviors (Szumlinski et al., 2004, 2005). Through their interactions with Homer proteins, the scaffolding proteins Shank and PSD-95 anchor Group 1 mGlu receptors within postsynaptic densities (Tu et al., 1999). Shank proteins are linked to NMDA receptors through PSD-95 associated complexes (Alagarsamy, Sorensen, & Conn, 2001; Xiao et al., 1998). In turn, Shank proteins enable mGlu1/5 receptors to associate and couple with NMDA receptor complexes (Bertaso et al., 2010). Analysis of the baseline expression of PSD-95 in male and female rats showed no sex differences in protein levels in the hippocampus, amygdala, or PFC (Wang et al., 2015). Taken together, the existing cell signaling literature has not identified major sex differences in trafficking or scaffolding of mGlu receptors.
2.3. Signaling and synaptic plasticity
Throughout the forebrain, mGlu receptors are generally conceptualized as modulatory components that regulate fast neurotransmission through glutamate or GABA receptors. Canonical signaling downstream of Group 1 mGlu receptors proceeds through activation of Gq proteins, phospholipase C (PLC), Protein Kinase C (PKC), and the release of intracellular calcium (Conn & Pin, 1997; De Blasi et al., 2001; Valenti, Conn, & Marino, 2002). In addition to PKC activation, Group 1 receptors can activate a myriad of downstream effectors, including the mitogen-activated protein kinase/extracellular receptor kinase (MAPK/ERK) pathway (Mao & Wang, 2016). mGlu1 and mGlu5 receptors can also facilitate the production of endocannabinoids, such as 2-arachadonylglycerol and anandamide (Ohno-Shosaku et al., 2002; Varma et al., 2001), which can attenuate pre-synaptic release probability through CB1 receptors and also regulate post-synaptic plasticity through the transient receptor potential cation channel TRPV1 (Chávez, Chiu, & Castillo, 2010; Grueter, Brasnjo, & Malenka, 2010). A recent study found that, in the rat PFC, 10Hz electrical stimulation evoked mGlu5 receptor-dependent LTD with distinct latent mechanisms across sexes (Bara et al., 2018). While LTD was mediated by mGlu5 receptors and endocannabinoid signaling in both sexes, LTD was blocked by the CB1 antagonists in male rats but TRPV1 antagonists blocked LTD in female rats. These findings highlight the tremendous need to revisit basic mechanisms to probe for potential latent or overt differences in mGlu receptor synaptic plasticity.
In addition to their ability to regulate synaptic plasticity, in some nuclei and cell types, mGlu1/5 receptor activation can elicit excitatory post-synaptic currents through more proximal actions on nonselective cation channels. For example, in several types of neurons within the cerebellum, mGlu1/5 receptor activation can elicit slow excitatory postsynaptic currents by activating transient receptor potential TRPC3 channels (Hartmann, Henning, & Konnerth, 2011). Interestingly, there are sex differences in cerebellar nuclei firing rates, with cerebellar nuclei cells from female mice having larger mGlu1/5 receptor currents than males, suggesting sex differences in mGlu1/5 receptor function (Mercer et al., 2016). However, there were no differences in Group 1 receptor protein levels between male and female mice, but blockade of glutamate transport increased the magnitude of mGlu1/5-dependent currents in male but not female mice. These data suggest that basal sex differences in cerebellar nuclei mGlu1/5 currents may not stem from receptor availability but are instead related to the access of glutamate to Group 1 receptors.
Group 2 and 3 mGlu receptors are generally linked to Gi/o proteins, which inhibit adenylyl cyclase to decrease cAMP production. Group 2 and 3 receptors can also initiate a variety of downstream signaling pathways through the liberation of Gβγ subunits, notably including the inhibition of calcium channels and facilitation of potassium channels. Similar to Group 1 receptors, Group 2 and 3 mGlu receptors can also modulate kinase signaling, such as MAPK and phosphatidylinositol 3-kinase (PI3K) pathways. Despite similarities in sequence homology and pharmacology, mGlu2 and mGlu3 receptors regulate neurotransmission through distinct mechanisms of action. While both Group 2 receptor subtypes act as presynaptic autoreceptors to reduce glutamate release probability, mGlu3 receptors are also heavily expressed at postsynaptic sites, where they can induce LTD through a postsynaptic signaling cascade involving mGlu5 receptors, Homer proteins, PI3K pathway signaling, and the internalization of AMPA receptors. Thus, any of the experience-dependent sex differences in mGlu5 receptor and/or Homer signaling (described in later sections), might conceivably have ramifications for mGlu3 receptor function as well.
2.4. Sex hormone regulation
Cellular pools of estrogen and other steroid hormones bind to both nuclear and membrane localized receptors, with varying affinity and response (Fuentes & Silveyra, 2019; Levin, 2009; Watson, Jeng, & Kochukov, 2008). In regions of the hippocampus, PFC, dorsal striatum, and nucleus accumbens, estrogen binds to membrane-localized receptors to activate second messenger systems and affect synaptic function (Milner et al., 2001; Razandi et al., 2004). Notably, several central effects of estradiol (E2) are linked to mGlu1/5 receptors. E2 alters neuronal excitability and synaptic plasticity through stimulation of Group 1 receptor machinery in both a sex-dependent and independent manner (McEwen et al., 2001; Tonn Eisinger et al., 2018). Membrane-localized estrogen receptors, estrogen receptor α (ERα) and ERβ, are functionally coupled to mGlu1/5 receptors expressed on the cell surface. Binding of E2 to these ER/mGlu complexes activates G proteins to trigger a glutamate-independent intracellular signaling cascade (Meitzen & Mermelstein, 2011). It is important to note that ER/mGlu complexes are present in both male and female brains, without major sex differences in overall expression. For example, immunocyto-chemistry studies targeting ERs in rat hippocampus (Weiland et al., 1997) and mouse amygdala (Bender et al., 2017) found no sex differences in ERα or ERβ expression levels. A more recent study separating mouse hippocampal ERs into nuclear and membrane populations detected greater extranuclear ERα in tissue from female mice, but no sex differences in ERβ expression (Mitterling et al., 2010). Moreover, ERα expression was highest in the CA1 region of mouse hippocampus during diestrus, while expression of ERβ in CA1 pyramidal cell layers was relatively high across the estrous cycle. We are not aware of any published findings describing Group 1 receptor expression throughout the estrous cycle.
Sex differences in ER/mGlu receptor localization and have been identified in the hippocampus. Co-immunoprecipitation studies indicate that ERα colocalizes with mGlu1 and mGlu5 receptors in both males and females (Boulware, Heisler, & Frick, 2013; Tabatadze et al., 2015). Interestingly, E2 treatment selectively increases the co-localization of ERα with mGlu1 receptors in females, without affecting co-localization with mGlu1 receptors in males or with mGlu5 receptors in either sex. Other studies report sex differences in expression of proteins associated with the organization and signaling of ERα/mGlur1 complexes. Transcripts for caveolin 1 and the palmitoylacyltransferase DHHC-7, were expressed at lower levels in the hippocampus of adult female rats compared to males (Meitzen et al., 2019), suggesting that posttranslational modifications may also guide sex differences in ERα/mGlu receptor co-localization. Interestingly, no sex differences in gene expression were detected in hippocampal samples from neonates, suggesting a possible developmental sex-dependent regulation of ERα/mGlu receptor complexes. Taken together, ER signaling promotes the surface expression of mGlu1/5 receptors to a greater extent in female individuals. Some evidence suggests the converse relationship holds as well (i.e., that mGlu receptors promote the expression of ERα). In ventromedial hypothalamus, genetic deletion of mGlu5 receptors decreased ERα expression in female mice but increased ERα expression in males (Fagan et al., 2020). In this study, mGlu5 receptors did not appear to regulate ERβ expression and mGlu1 receptor function was not addressed. Taken together, the bidirectional relationship between ERα and mGlu1/5 receptors raises the possibility that sex differences in downstream signaling may govern mGlu1/5 receptor localization and function.
There are both sex and region differences in the pairing of ER/mGlu receptor subtype coupling and their downstream effects. E2 facilitates IP3 production in hippocampal slices from female rats to a much greater extent than in male rats (Tabatadze et al., 2015). E2-driven IP3 production was abolished by JNJ16259685, indicating an mGlu1 receptor-dependent mechanism. Importantly, no difference in IP3 production was observed during pharmacological activation of mGlu1 with the Group 1 agonist DHPG (Tabatadze et al., 2015), suggesting that the sex differences in signaling is manifested upstream of mGlu receptors. Other studies have yielded similar results and identified additional signaling nodes. In hippocampal cultures from female rats, E2 facilitates CREB phosphorylation through a pathway downstream of mGlu1 receptors, PLC, PKC, IP3 receptors, and MAPK (Boulware et al., 2005). In these studies, an mGlu5 negative allosteric modulator (NAM) had no effect, again indicating specific coupling between ERα and mGlu1 receptors. Furthermore, ERα/mGlu1 receptor-dependent CREB phosphorylation was specific to female hippocampal cultures, whereas similar E2 treatment in male cultures had no effect on CREB phosphorylation (Boulware et al., 2005). Together, these studies indicate that E2 signals through mGlu1 receptor and canonical Gq signaling pathways in the female hippocampus. By contrast, E2 activates mGlu5 receptor signaling in striatum. In female striatal cultures, the rapid actions of E2 lead to MAPK-dependent CREB phosphorylation through the activation of ERα/mGlu5 receptor signaling complexes (Grove-Strawser, Boulware, & Mermelstein, 2010). Similar to the studies in hippocampus cultures, male striatal cultures did not respond to E2 treatment. Thus, while ER-mGlu1/5 receptor interactions are potentiated in the brains of female individuals, the mGlu receptor subtype involved varies based on region.
Functional studies also support the conclusion that there are sex differences in ERα/mGlu1/5 receptor regulation of neuronal physiology and synaptic plasticity. In ventromedial hypothalamus, genetic deletion of mGlu5 receptors decreases current-evoked spike-firing in female but not male mice (Fagan et al., 2020). These actions were linked with E2 signaling, as the decreased excitability in mGlu5 receptor knockouts was occluded by ovariectomy in control mice and restored by E2. Neurons within cerebellar nuclei also display larger mGlu1/5 slow excitatory currents in female mice relative to males (Mercer et al., 2016). In addition to basic physiology and excitability, a growing literature now indicates that gonadal hormones regulate mGlu1/5 receptor-dependent synaptic plasticity. While E2 is synthesized in the hippocampus of both males and females, the hormone exerts sex-specific effects on synaptic plasticity in CA1. In female hippocampal pyramidal cells, E2 suppresses inhibitory synapses through activation of post-synaptic ERα/mGlu receptor complexes, retrograde signaling through anandamide and CB1 receptors, and the attenuation of GABA release probability (Huang & Woolley, 2012). E2 also potentiates excitatory transmission in hippocampus through distinct mechanisms of action in females and males (Oberlander & Woolley, 2016), but it remains unclear whether mGlu receptor signaling is involved in these phenomena.
Naturally occurring fluctuations in apical dendritic spine density of CA1 hippocampal pyramidal cells are mediated, in part, by fluctuating E2 levels across the estrous cycle in female rats (Woolley et al., 1990; Woolley & McEwen, 1993). Studies in nucleus accumbens suggest that these endogenous cycles are regulated by mGlu1 and mGlu5. Consistent with the results from functional studies, E2 and other gonadal hormones alter dendritic spine plasticity in medium spiny neurons of the nucleus accumbens through the activation of Group 1 receptors (Gross et al., 2016, 2018). Though mGlu1/5 receptors are structurally similar and tend to signal through conserved downstream pathways, evidence suggests that mGlu1 and mGlu5 receptors regulate spine density through distinct mechanisms. For example, systemic administration of selective mGlu1 and mGlu5 positive allosteric modulators increase and decrease spine density, respectively, in the nucleus accumbens of female rats (Gross et al., 2016). In addition, this structural plasticity is linked to gonadal hormones. For instance, E2 decreases spine density in nucleus accumbens core medium spiny neurons, but not dorsal striatum, and this effect was blocked by MPEP (Peterson, Mermelstein, & Meisel, 2015). Similar to E2, the male hormone dihydrotestosterone (DHT) can also influence dendritic plasticity through Group 1 activation. In castrated males, DHT administration decreased dendritic spine density on MSNs in the nucleus accumbens shell, but not core, of castrated male rats and this effect was blocked by pretreatment with MPEP (Gross et al., 2018). Thus, similar to their dissociable roles in mediating gonadal hormone effects on neuronal physiology, mGlu1, and mGlu5 receptors differentially regulate hormonal effects on structural plasticity in a region-specific manner.
Group 2 receptor signaling and female sex hormone signaling converge in the regulation of ion channels and activity-dependent gene transcription. Initial studies independently show that Group 2 receptor agonists (Chavis et al., 1994) and E2 each lead to the inhibition of L-type calcium channel currents through either ERα or ERβ (Chaban et al., 2003; Mermelstein, Becker, & Surmeier, 1996). LY341495 (at concentrations selective for mGlu2/3 receptors) blocks the ability of E2 to attenuate calcium channel currents and CREB phosphorylation (Boulware et al., 2005). Though the mGlu2/3 agonists suppressed CREB phosphorylation similarly in hippocampal cultures from male rats, E2’s effects were specific to cultures from female rats. Additional studies have begun to tease apart the relative contribution of mGlu2 vs mGlu3 receptors. In striatal cultures from female rats, studies using subtype-specific interfering RNA revealed that knockdown of mGlu3 receptors, but not mGlu2 receptors, prevented E2 from attenuating CREB phosphorylation (Grove-Strawser et al., 2010). In addition, neither mGlu2 nor mGlu3 receptor expression appears to be altered by estrus cycle. Comparable receptor levels were detected in the nucleus accumbens when cocaine/vehicle self-administration rats were killed in the estrus vs. non-estrus phases (Logan et al., 2020). It is important to note, however, that these findings may or may not generalize to other brain areas.
3. Insight gained from behavioral pharmacology, transgenic mice, and disease models
Pharmacological and genetic manipulation of mGlu receptor subtypes in preclinical research have revealed further insights into their biological functions. Over the past two decades, numerous studies have used highly selective pharmacological ligands and transgenic animals to characterize the function of mGlu receptor functions in male preclinical models (each of which have been extensively detailed in (Niswender & Conn, 2010)). In stark comparison, only a handful of studies have addressed mGlu1/5 receptor function in females, and even fewer studies have made direct comparisons to probe for potential sex differences. Nonetheless, the results of those few studies are consistent with significant sex differences in mGlu receptor functions in basal and altered states.
3.1. Cognition
mGlu1 (Gil-Sanz et al., 2008) and mGlu5 (Simonyi, Schachtman, & Christoffersen, 2005; Xu et al., 2009) receptors have been widely implicated in associative learning based on studies performed in male rodents (Luscher & Huber, 2010). However, it remains unclear whether similar contributions of mGlu1 and mGlu5 receptors guide cognition in female individuals. In a study using mice with selective deletion of mGlu5 receptors from parvalbumin-expressing interneurons, there was a sex difference in perseverative behavior on the Barnes maze (Barnes et al., 2015). Male mice lacking mGlu5 receptors in parvalbumin cells took longer to escape the Barnes maze and displayed more preservative errors than controls. By contrast, while female knockout mice also performed worse than female controls, changes in perseveration were not closely tied to the cognitive disruption. In addition, a genotype by sex interaction was observed with respect to auditory event related potentials, suggesting that mGlu5 receptor signaling in cortical parvalbumin interneurons may differentially regulate rhythmic activity and behavioral flexibility across sexes. Considering that other studies observed no differences in PFC-dependent cognitive behaviors in models where mGlu5 receptor function on pyramidal cells was disrupted (Bara et al., 2018), these findings reinforce that mGlu receptor function can regulate behavior in a cell type-specific manner.
Studies investigating sex hormone regulation of learning and memory also provide clear indications that mGlu1/5 receptor signaling may guide latent sex differences in the molecular mechanisms of cognition. For example, E2 delivery to the dorsal hippocampus of female mice enhances novel object recognition (Fernandez et al., 2008; Zhao et al., 2012; Zhao, Fan, & Frick, 2010), an effect blocked by co-administration of LY367385 or the MEK inhibitor U0126 (Boulware et al., 2013). Considering that the female hippocampus displays enhanced ERK-dependent ER/mGlu1 receptor signaling (see earlier section), a compelling hypothesis for future research is that mGlu1 receptors differentially contribute to hippocampal-dependent cognition across sexes. Similarly, some evidence suggests sex-differences in the contribution upregulation of hippocampal mGlu5 receptors to cognitive processing. In a model of phenylketonuria, male but not female mice, displayed increased mGlu5 receptor protein levels in the hippocampus that coincided with a deficit in object location recognition (Nardecchia et al., 2018). The increased mGlu5 receptor expression occurred at the protein, but not transcript, level and was also associated with increased expression of long Homer isoforms. Additionally, administration of MPEP to mutant male mice significantly improved behavioral abnormalities. Taken together, the preclinical literature suggests that potential mGlu1 and mGlu5 modulators may display sex differences in pro-cognitive efficacy.
Genetic deletion of mGlu8 receptors alters hippocampal-dependent learning and memory (Duvoisin et al., 2010). Though mGlu8 receptor deletion impaired novel object recognition and spatial memory retention in the Morris water maze in both males and females, the effects on memory retention were more pronounced in females. Later studies from the same group found impaired contextual fear learning in female knockout mice relative to wildtypes, whereas males were not affected (Torres et al., 2018). Together, this literature raises the possibility that hippocampal circuits in female individuals are particularly sensitive to manipulations in mGlu8 receptor function. Transgenic mouse studies have also identified that mGlu7 receptors are involved in working memory (Hölscher et al., 2004), fear and extinction learning (Goddyn et al., 2008), anxiety-like behaviors (Cryan et al., 2003), and social behaviors (Fisher et al., 2020, 2021). In each of these studies, female mice and male mice were included, similar effects were observed across sexes, and the datasets were collapsed for subsequent analysis and presentation. Thus, there is minimal evidence to suggest notable sex differences in mGlu7 receptor function regulate cognition or other behaviors.
3.2. Social behaviors
As mentioned previously, Group 1 receptors drive endocannabinoid signaling to regulate synaptic plasticity and behavior (Varma et al., 2001). Previous studies involving prenatal cannabinoid exposure have linked adaptations to PFC mGlu1/5 receptor function and endocannabinoids to social behavior in a sex-dependent manner. WIN-55,212 administration during gestation causes decreases in adult social exploration and play behaviors in male but not female rats (Bara et al., 2018). There were no differences in the elevated plus maze or in an order recognition task following gestational cannabinoid exposure, suggesting changes were specific to social behaviors in male individuals. In addition, prenatal cannabinoid exposure eliminated endocannabinoid LTD induced by 10-Hz stimulation in layer 5 pyramidal neurons of the PFC in male but not female rats. Previous studies have found this 10-Hz LTD is mediated by mGlu5 receptors (Luscher & Huber, 2010), and the mGlu5 PAM CDPPB restored LTD in male rats exposure to prenatal cannabinoids (Bara et al., 2018). Finally, the authors linked the sex-specificity of these phenomena to the latent mechanisms mediating PFC LTD, providing evidence that LTD in male rats is mediated by CB1 receptors while TRPV1 receptors mediate LTD in female rats (Bara et al., 2018). Systemic treatment with either the mGlu5 PAM or a fatty acid amide hydrolase inhibitor URB597 rescued behavioral deficits in affected male rats, providing a sex-dependent link between mGlu5 receptor plasticity and social interaction.
Evidence linking mGlu5 receptor function with social behaviors has also emerged from transgenic animal studies investigating Shank3. Deficits in Shank3, a scaffolding protein that coordinates mGlu5/Homer intracellular signaling, are associated with the pathogenesis of autism spectrum disorder (Uchino & Waga, 2013; Verpelli et al., 2011; Zoicas & Kornhuber, 2019). Mutant Shank3 mice exhibit significant impairments in social behaviors and hippocampal mGlu5 receptor signaling, without obvious sex differences (Matas et al., 2021; Peixoto et al., 2016; Wang et al., 2011). While social behaviors related to Shank3 disruption appear comparable in both sexes, Shank3 mutant mice also exhibit changes in repetitive behaviors and motor control, with evidence of sex differences in effects. Male Shank3 mutants displayed greater impairments in several gait parameters than littermate female mutants (Wang et al., 2011), and this phenotype has been linked with lower expression of mGlu5 receptors in cerebellum in male mice (Matas et al., 2021). Together, these studies suggest that Shank3 regulation of mGlu5 receptor function exhibits sex differences in specific brain areas (i.e., cerebellum but not hippocampus) and indicate that not all insults to mGlu5 receptor signaling affect social play in a sex-dependent manner.
3.3. Alcohol use
Genetic and pharmacological manipulations of mGlu1/5 receptors have revealed a clear association with alcohol seeking behaviors (Joffe et al., 2018). The link between ethanol exposure and mGlu1/5 receptor function is bidirectional. Acute and chronic applications of ethanol alter mGlu1/5 receptor function in cultured oocytes and rat cerebellar slices (Belmeguenai et al., 2008; Carta, Mameli, & Valenzuela, 2006; Minami et al., 1998; Netzeband & Gruol, 1995). On the other hand, Group 1 antagonists, such as the prototypical mGlu5 NAM MPEP, reduce ethanol-seeking and relapse-like behaviors in a broad variety of models and background (Backstrom et al., 2004; Besheer et al., 2008; Hodge et al., 2006; Lominac et al., 2006). While most of these studies were limited to male subjects, a few have found comparable effects in females. In a recent study directly assessing sex differences in binge drinking, MPEP displayed a similar dose-response curve in reducing binge alcohol consumption in male and female mice (Huang, Thompson, & Taylor, 2021). Similarly, MTEP administration decreased binge ethanol intake in male and female mice (Cozzoli et al., 2014). Interestingly, in that study, female but not male mice displayed increased ethanol consumption 24 h after MTEP exposure, suggesting that females may be uniquely sensitive to compensatory or rebound adaptations following mGlu5 inhibition.
Alterations in mGlu1/5 receptor signaling in several brain areas have been implicated in binge drinking models. Several studies have implicated Homer2 in the regulation of ethanol sensitivity and ethanol-induced synaptic plasticity (Szumlinski et al., 2005, 2008; Szumlinski, Ary, & Lominac, 2008), including initial studies using mixed-sex cohorts. Homer2 knockout mice of both sexes resist drinking high concentrations of ethanol and do not express ethanol locomotor sensitization or conditioned place preference. By contrast, binge-drinking upregulates mGlu5 receptors, Homer2, and PI3K signaling in male mice (Cozzoli et al., 2009). Furthermore, both male and female transgenic mice that express a point mutation in mGlu5 receptors rendering them insensitive to ERK phosphorylation exhibit decreased sensitivity to the aversive properties of high dose ethanol (Campbell et al., 2019). Taken together, this literature indicates that upregulation of Homer2/mGlu5 receptor expression and activation of ERK-dependent phosphorylation function as adaptive compensatory mechanisms to limit alcohol consumption in both sexes.
Women are more likely than men to drink alcohol to regulate negative emotional state or stress (Peltier et al., 2019). In recent years, the sexually dimorphic bed nucleus of the stria terminalis (BNST) has been identified as a point of convergence for early stress- and alcohol-related disorders in modulating negative affect behaviors (Kasten et al., 2021; Lebow & Chen, 2016). This observation from AUD has been modeled by inducing dependence to ethanol during adolescence in rodents (Crews et al., 2016). In adolescent female mice, acute withdrawal from chronic intermittent ethanol disrupted mGlu5 receptor LTD in the BNST (Kasten et al., 2020). By contrast, male mice in acute withdrawal exhibited no differences in mGlu5 receptor LTD, but instead displayed enhanced NMDA receptor functions (Carzoli et al., 2019). Interestingly, these sex-dependent changes in BNST plasticity mapped onto distinct behavioral phenotypes associated with withdrawal. Adolescent female mice showed enhanced anxiety/anhedonia-like behavior in a novelty-induced hypophagia task, whereas male mice in withdrawal displayed elevated freezing during fear conditioning paradigms (Kasten et al., 2020). Two pieces of convergent evidence link the adaptations to BNST mGlu5 receptor plasticity with female-specific affective behaviors. First, systemic delivery of MTEP to female mice reversed withdrawal-induced increases in latency to each in the novelty-induced hypophagia assay (Kasten et al., 2020). Second, mGlu5 receptor knockdown in the BNST reproduces anhedonia-like behavior in the novelty-induced hypophagia task in female but not male mice (Kasten et al., 2021). Consistent with the findings by Kasten et al. (2020) in mice, Chandler et al. also observed that adolescent chronic intermittent ethanol exposure enhanced contextual fear learning and spontaneous recovery of freezing behavior in male but not female rats (Chandler, Vaughan, & Gass, 2022). Interestingly, the mGlu5 PAM CDPPB reversed the withdrawal-induced phenotypes in male rats, although it is not clear from these studies how female individuals would respond to enhanced mGlu5 receptor function. Together, these findings indicate that mGlu5 receptors influence behavioral phenotypes relevant to alcohol dependence in adolescence in both males and females, however there is a clear impact of sex on resulting behaviors (Table 1).
Table 1.
Sex differences in mGlu receptor regulation of affective behaviors.
Domain | Family | Subtype | Species/Strain | Sex Diff | Key findings | First author (year) |
---|---|---|---|---|---|---|
Anxiety-like and affective behavior | Group I | mGlu1/5 | Rat, SD | Y | BLA infusion of the mGlu1/5 agonist DHPG increased punished drinking in VCT in OVX ♀. BLA infusion of the DHPG decreased punished drinking in VCT in ♂ (De Jesús-Burgos, Torres-Llenza, & Pérez-Acevedo, 2012) |
De Jesús-Burgos (2016) |
mGlu5 | Mouse, C57BL/6J, Grm5(AA/AA) | N | Mice with mGlu5 that cannot be phosphorylated by ERK displayed decreased float time in FST and increased open arm time in the EPM (Cozzoli et al., 2009) | Campbell (2019) | ||
Mouse, C57BL/6J, Grm5loxp | Y | mGlu5 KD in BNST prolonged latency to eat in NIH in ♀ but not ♂. mGlu5 KD in BNST decreased center time in open field in ♂ but not ♀ (Lebow & Chen, 2016) |
Kasten (2021) | |||
Mouse, C57BL/6J, Grm5loxp, SST-Cre | N | SST-mGlu5-KO mice resisted stress-induced alterations in PFC inhibitory transmission and spontaneous alternation on the Y maze (Di Menna et al., 2018) | Joffe (2022) | |||
Mouse, C57BL/6J, Grm5loxp, PV-Cre | Y | ♂ PV-mGlu5-KO mice displayed more perseverative errors on the Barnes maze. ♀ KO mice did not display more perseverative errors than controls (Luscher & Huber, 2010) |
Barnes (2015) | |||
Group 2 | mGlu3 | Mouse, C57BL6/J x C57BL6/Ntac, Grm3loxp | N | mGlu3 KD in PFC increased open arm time in EZM and decreased immobility in TST/FST (Maksymetz & Joffe, 2021) | Joffe (2020) | |
Group 3 | mGlu4 | Mouse, C57BL6/J, Grm4−/− | Y | ♀ mGlu4 KO mice displayed decreased open arm/center time in EZM and open field. ♂ mGlu4 KO mice displayed increased open arm/center time in EZM and open field (Joffe et al., 2021) |
Davis (2012) | |
mGlu8 | Mouse, C57BL6/J, Grm8−/− | Y | ♀ mGlu8 KO mice displayed increased open arm/center time in EZM and open field, and increased startle response. ♂ mGlu8 KO mice displayed decreased open arm/center time in EZM and open field, and increased startle response. (Joffe et al., 2021) |
Duvoisin (2010) | ||
Alcohol Use | Group I | mGlu5 | Mouse, C57BL/6J | Y | Acute withdrawal from AIE disrupts mGlu5 LTD in the BNST in ♀ but not ♂ (Crews et al., 2016) | Kasten (2020) |
Mouse, C57BL/6NCrl | N | mGlu5 NAM MPEP (30mg/kg) decreases binge drinking (Besheer et al., 2008) | Huang (2021) | |||
Mouse, C57BL/6J | Y | mGlu5 NAM MTEP (20 mg/kg) decreases binge drinking. One month later, increased drinking was observed in ♀ but not ♂ (Huang et al., 2021) |
Cozzoli (2014) | |||
Mouse, C57BL/6J, Grm5(AA/AA) | N | Transgenic mice with mGlu5 that cannot be phosphorylated by ERK display increased drinking (Cozzoli et al., 2009) | Campbell (2019) |
All studies in this table employed mixed-sex cohorts
AIE, adolescent intermittent ethanol vapor; BLA, basolateral amygdala; BNST, bed nucleus of the stria terminalis; EPM, elevated plus-maze; ERK, extracellular signal-regulated kinase; EZM, elevated zero-maze; FST, forced swim test; KD, knock-down; KO, knock-out; LTD, long-term depression; NAM, negative allosteric modulator; OVX, ovariectomized; PFC, prefrontal cortex; PV, parvalbumin; SD, Sprague Dawley; SST, somatostatin; TST, tail suspension test; VCT, Vogel conflict test.
3.4. Drug use
Compared to males, female rodents acquire drug self-administration much more quickly, rapidly progress through milestones of drug use, and exhibit greater levels of drug reinstatement, mediated in part by the estrous cycle (Anker & Carroll, 2011; Carroll et al., 2002; Doncheck et al., 2018; Feltenstein & See, 2007). A significant literature, albeit generally restricted to males, indicates that Group 1 mGlu receptors contribute to drug-seeking and related behaviors. In general, mGlu1 antagonists and NAMs block some cocaine-seeking behaviors (Xie et al., 2010; Yu et al., 2013), whereas mGlu1 PAMs may prevent relapse-like behaviors (Loweth et al., 2014). While these studies allude to the involvement mGlu1 receptors in the adaptations to chronic drug use, there is still limited information on how sex may influence mGlu1 activity in SUD. In comparison to mGlu1, we have a better understanding how sex differences in mGlu5 receptor signaling guide behaviors related to drug use. Some of this evidence comes from studies examining sex hormone contributions to drug-seeking. In female individuals, the subjective effects of smoked cocaine fluctuate over the course of the menstrual cycle (Evans, Haney, & Foltin, 2002). Similar findings have been observed in several rodent models and laboratories: female rodents show increased cocaine-induced hyperlocomotion, enhanced cocaine cue and reward learning, increased motivation to self-administer, increased incubation of craving, and estrous cycle-dependent cocaine reinforcement (Calipari et al., 2017; Jackson, Robinson, & Becker, 2006; Johnson et al., 2019; Roberts, Bennett, & Vickers, 1989). In addition, in female rodent ovariectomy studies, E2 treatment facilitates cocaine self-administration (Hu & Becker, 2003; Lynch et al., 2001), related effects on hyperlocomotion and self-administration were blocked by MPEP administration (Gross et al., 2016; Martinez et al., 2014). Taken together, this literature suggests that changes in drug-seeking and cue attending during female hormonal cycles may be dependent on surges in E2 and potentiated signaling through downstream mGlu5 receptors (Table 2).
Table 2.
Sex hormone regulation of mGlu receptor synaptic plasticity.
Family | Subtype | Region | Species/Strain | Sexes used | Sex diff | Key findings | First author (Year) |
---|---|---|---|---|---|---|---|
Group I | mGlu1 | HPC | Rata | Both | Y | E2 facilitates CREB phosphorylation through ERα/mGlu1, PLC, PKC, and IP3R in ♀ but not ♂ (Boulware et al., 2005) | Boulware (2005) |
Rat, SD | Both (OVX) | Y | E2 suppresses GABA release probability through ERα/mGlu1, anandamide, and CB1R in ♀ but not ♂ (Huang & Woolley, 2012) | Huang (2012) | |||
Rat, SD | Both (OVX/CAS) | Y | E2 suppression of GABA release probability coincides with greater IP3 production in ♀ than ♂. ERα/mGlu1 complexes in exist in ♂ but are only regulated by E2 in ♀ (Tabatadze et al., 2015) | Tabatadze (2015) | |||
Rat, SD | ♀ (OVX) | n.a. | E2 increases dendritic spine density through activation of mGlu1 (Peterson et al., 2015) | Peterson (2015) | |||
mGlu5 | Striatum | Rata | ♀ | n.a. | E2 facilitates CREB phosphorylation through ERα/mGlu5 and MAPK (Grove-Strawser et al., 2010) | Grove-Strawser (2010) | |
NAC core | Rat, SD | ♀ (ovx) | n.a. | E2 decreases dendritic spine density through activation of mGlu5 (Peterson et al., 2015) | Peterson (2015) | ||
NAC shell | Rat, SD | ♂ (CAS) | n.a. | DHT decreases dendritic spine density in the NAC shell, but not core, through mGlu5 (Gross et al., 2018) | Gross (2018) | ||
Group II | mGlu2/3 | HPC | Rata | Both | Y | E2 decreases CREB phosphorylation through mGlu2 and/or mGlu3 inhibition of L-type calcium channels in ♀ but not ♂ (Boulware et al., 2005) | Boulware (2005) |
mGlu3 | Striatum | Rata | ♀ | n.a. | E2 decreases CREB phosphorylation through mGlu3 inhibition of L-type calcium channels (Grove-Strawser et al., 2010) | Grove-Strawser (2010) |
Pyramidal neurons cultured from P1-P2 rat pups, experiments performed at 9 d.i.v.; strain not specified
CAS, castrated; CB1R, cannabinoid receptor type 1; CREB, cAMP response element-binding protein; DHT, dihydrotestosterone; GABA, gamma-aminobutyric acid; E2, estradiol; ER, estrogen receptor; HPC, hippocampus; IP3R, inositol triphosphate receptor; MAPK, mitogen-activated protein kinase; n.a., not addressed; NAC, nucleus accumbens; OVX, ovariectomized; PLC, phospholipase C; PKC, protein kinase C; SD, Sprague Dawley
3.5. Anxiety-like and affective behavior
Affective disorders, such as major depressive disorder and anxiety-based disorders, including post-traumatic stress and generalized anxiety disorders, are more prevalent in women than in men (Altemus, Sarvaiya, & Neill Epperson, 2014; Bangasser & Cuarenta, 2021; Kessler et al., 2012). Though monoaminergic systems have historically been the predominant focus for therapeutic targets, growing evidence suggests a functional role for the glutamate system in the treatment of anxiety and affective disorders (Joffe et al., 2019; Mathews, Henter, & Zarate, 2012; Palucha & Pilc, 2007). Indeed, several studies have reported the robust anxiolytic-like effects in preclinical models following pharmacological manipulations of Group 1 receptors (Ballard et al., 2005; Busse et al., 2004; Pietraszek et al., 2005), although this literature is heavily biased toward male subjects.
Stress, a major contributing factor to the development of anxiety and affective disorders, dynamically alters Group 1 receptor function. In the PFC of male rats, exposure to 2-days forced swim stress decreased mGlu1 and increased mGlu5 receptor protein expression (Wang et al., 2015). Consistent with a stress-induced dysregulation of PFC mGlu5 receptor signaling, 20-min of restraint stress in male mice impairs the induction of mGlu3 LTD that is dependent on mGlu5 receptor signaling (Di Menna et al., 2018; Joffe et al., 2019). By contrast, female rats exposed to forced swim stress displayed intact mGlu1 and decreased mGlu5 receptor expression (Wang et al., 2015). Continued research should account for cell type-specific differences in mGlu5 receptor signaling, as we recently found that restraint stress potentiates somatostatin interneuron activity through a mGlu5 receptor-dependent mechanism in both male and female mice (Joffe et al., 2022). Together, these results suggest that stress-induced alterations in mGlu1/5 receptor signaling may mediate some effects of stress and potentially increase vulnerability to affective behaviors in a sex-specific manner.
In female rodents, E2 can reduce anxiety-like behaviors (Hill, Karacabeyli, & Gorzalka, 2007; Mora, Dussaubat, & Díaz-Véliz, 1996); these effects are mediated by activation of Group 1 mGlu receptors (De Jesús-Burgos et al., 2012) and endocannabinoid mobilization (Hill et al., 2007). Studies from ovariectomized female rats suggest that the ability of mGlu1/5 receptors to regulate anxiety-like behavior may vary across the estrous cycle. In E2-treated ovariectomized female rats, direct infusion of DHPG into the basolateral amygdala decreased anxiety-related behaviors in the elevated plus maze (De Jesús-Burgos et al., 2012) and Vogel conflict tests (De Jesús-Burgos et al., 2016). By contrast, this manipulation had no effect on non-E2 treated ovariectomized females in either assay or in males in the plus maze. Furthermore, stimulation of mGlu1/5 receptors within the BLA increased anxiety-like behavior in male rats on the Vogel conflict test (De Jesús-Burgos et al., 2016). It is tempting to speculate that these differences could be related to differential contributions of CB1 receptor signaling to anxiety-like behavior (Bowers & Ressler, 2016), but more studies are needed to fully evaluate that hypothesis. In any case, the results from these studies are consistent with a sex-specific role of amygdalar Group 1 receptors in the attenuation of anxiety and depressive-type behaviors.
Antidepressant-like and anxiolytic-like phenotypes have been observed in mGlu2 and mGlu3 receptor knockout mice and following administration of compounds that inhibit mGlu2 and/or mGlu3 receptors (Highland et al., 2019; Joffe et al., 2020; Maksymetz & Joffe, 2021). Unfortunately, most of these studies were restricted to male mice. One exception is our recent study reporting a panel of emotional behavioral studies assessing frontal cortex deletion of mGlu3 receptors (Joffe et al., 2021). We observed that frontal cortex knockdown of mGlu3 receptors decreased immobility in the forced swim and tail suspension tests and increased open-arm time in the elevated-zero maze. We found no evidence for an interaction between mGlu3 receptor knockout and sex; however, it is unclear to what extent these findings may generalize to additional behaviors, mGlu3 receptor function in other brain areas, or acute pharmacological receptor inhibition. Additional studies examining potential sex-dependent behavioral effects of mGlu2 and mGlu3 modulators are certainly warranted.
Studies using selective pharmacology and knockout mice implicate Group 3 receptors in anxiety-like behaviors. Systemic treatment with the mGlu4 PAM ADX88178 increased open-arm entries on the elevated plus maze in both male and female mice (Kalinichev et al., 2014), suggesting that mGlu4 receptor signaling can reduce anxiety-like behavior in both sexes. Consistent with this hypothesis, male mGlu4 receptor knockout mice display increased anxiety-like behavior in the open field and elevated plus maze that emerge at age 12 months (Davis et al., 2012). In striking contrast, however, female mGlu4 receptor knockouts display reduced anxiety-like behaviors in both assays at 6 and 12 months. The authors observed similar sex- and age-dependent effects on sensorimotor function assessed on the accelerating rotarod. Male knockout mice at 12 months, but not 6 months, fell from the rotarod more quickly than wild-type controls, whereas female knockout mice at both ages displayed longer latencies to fall than female wildtypes. Interestingly, in the same studies, mGlu4 knockout mice displayed altered cued fear learning across all ages and sexes. Thus, the sex- and age-dependencies of mGlu4 receptor regulation of anxiety-like and sensorimotor behaviors likely occur through actions within one or more distinct neural circuits, rather than at the cellular level of receptor expression or function. Considering that mGlu4 receptors are highly expressed in corticostriatal and thalamostriatal circuits—which are heavily implicated in anxiety and motor function but less so in fear learning—it is tempting to speculate that these receptor populations contribute to the observed sex- and age-dependent behavioral phenotypes in mGlu4 knockout mice. Clearly, additional studies will be needed to address this speculative hypothesis.
Numerous studies suggest that mGlu4 and mGlu8 may have related and overlapping functions, particularly with respect to anxiety-like behaviors. Similar to the sex-dependent findings in mGlu4 receptor knockout mice, there is a sex difference in anxiety-like behavior in mGlu8 receptor knockouts. In the elevated plus maze, open-arm time and entries were decreased in male knockouts but increased in female knockouts, respectively suggesting enhanced and reduced anxiety-like behaviors (Duvoisin et al., 2010). Consistent with this finding, the acoustic startle response was enhanced in male knockout mice but attenuated in female knockouts. An important caveat to these findings, however, is that mGlu8 knockouts of both sexes exhibited less time in the center of an open field relative to matched controls. Therefore, the broad generalizability of the anxiety-like phenotype of female knockout mice should be interpreted with some caution.
3.6. Neurodegeneration
Alzheimer’s disease, a type of dementia, is characterized as a progressive neurological disorder in which atrophy of the brain leads to cell death and subsequent loss of cognitive functions (Johnson et al., 2019). Though the incidence of Alzheimer’s disease does not differ by sex, there are distinct sex-differences in the symptomatology, progression, and treatment of Alzheimer’s disease (Ferretti et al., 2018). Aggregation of β-amyloid proteins in the formation of amyloid plaques is a pathological hallmark of Alzheimer’s disease in both sexes (Murphy & LeVine 3rd, 2010). In fact, accumulation of oligomers induces abnormal clustering of mGlu5 receptors at the cell membrane and, in turn, alters mGlu5 glutamate transmission leading to synaptic degradation (Renner et al., 2010). In cortical tissue of wild-type male mice and human men, β-amyloid oligomers display a high-affinity (nanomolar range) interaction with mGlu5 receptors, an effect not observed in female mice or in women (Abd-Elrahman et al., 2020; Abd-Elrahman & Ferguson, 2022). Furthermore, in primary neuronal cultures from male but not female mice, β-amyloid oligomers activate glycogen synthase kinase 3β signaling in an mGlu5 receptor dependent manner. Consistent with these actions, systemic treatment with CTEP mGlu5 inhibition reduced β-amyloid pathology and improved object recognition in male disease model mice but had no effect in females (Abd-Elrahman et al., 2020; Abd-Elrahman & Ferguson, 2022). An mGlu5 silent allosteric modulator reversed deficits in hippocampal-dependent cognition in male Alzheimer’s disease model mice, but cohorts of female mice were not included in that study (Haas et al., 2017). Taken together, these findings suggest a male-specific contribution of mGlu5 receptor signaling in β-amyloid-driven pathology in Alzheimer’s disease.
Evidence from Huntington’s disease models similarly suggests that male individuals may be more sensitive to neurodegeneration related to mGlu5 receptor signaling. MPEP administration in male mouse models of Huntington’s disease inhibits disease progression (Schiefer et al., 2004) and mGlu5 knockout improves motor behavior and decreases aggregate size of mutant huntingtin (Ribeiro et al., 2014). There is growing preclinical and clinical evidence to suggest that sex may influence the phenotype of Huntington’s disease (Bode et al., 2008; Cao et al., 2019; Zielonka et al., 2013), and preclinical models are beginning to take this variation into account. A recent study examined CTEP administration in male and female Huntington’s disease model mice; although both sexes displayed improvements in HD neuropathology following mGlu5 blockage, female HD mice required longer CTEP treatment to show motor improvements (Li et al., 2022). Furthermore, mGlu5 antagonism did not improve cognitive impairments or grip strength in female HD mice. These findings collectively suggest that males may be more sensitive to neurodegenerative processes related to aberrant mGlu5 receptor signaling. One intriguing possible mechanism could be sex differences in coordination between mGlu3 and mGlu5 receptor signaling, as the same group observed that LY379268 exerts sex-dependent effects on Akt, GSK3β, and ERK1/2 phosphorylation in the striatum of Huntington’s disease model mice (Li et al., 2022).
4. Conclusions
Despite preclinical evidence to suggest possible sex-dependent differences in mGlu receptor expression and function, few human studies have been adequately powered to make well-justified conclusions and it has been challenging to make clear comparisons across species. For example, positron emission tomography has enabled the quantification of available mGlu5 receptors in the intact human brain, and the allosteric radiotracer [11C]-ABP688 has been used to assess sex differences in mGlu5 receptor availability. While sex differences in mGlu5 receptor availability were not detected in a small study (DuBois et al., 2016), a larger study found significantly higher rates of [11C]-ABP688 binding potential in men than women across several regions of the PFC, striatum, and hippocampus (Smart et al., 2019). The largest magnitude differences were observed in the orbitofrontal cortex and the dorsolateral PFC, in which binding potentials were 22% and 20% greater in men, respectively. Overall, a whole-brain comparison of mGlu5 receptor availability found that [11C]-ABP688 binding potential was 17% greater in men. Consistent with these findings, an earlier human PET imaging study using [11C]-ABP688 reported 31% greater global mGlu5 binding potential in non-smoking men when compared to non-smoking women (Akkus et al., 2013). Taken together, the larger availability of mGlu5 receptors in women compared to men seems to conflict with the preclinical literature, as major sex differences in mGlu5 receptor expression have not been consistently reported in rodents (Wickens et al., 2018) and available evidence points toward decreased cortical mGlu5 receptor expression in male rats compared to females (Wang et al., 2015). Importantly, technical considerations impede direct comparisons between the clinical and preclinical literature. For one, studies using radioligands like [11C]-ABP688 detect mGlu5 receptors available on the cell surface, while Western blots with targeted antibodies can detect receptor populations in intracellular (Purgert et al., 2014) or nuclear (Jong et al., 2005) membranes. In addition, mGlu receptor antibodies were raised against sections of the intracellular C-terminal tail or the N-terminal extracellular domain, whereas allosteric ligands like [11C]-ABP688 interact with the allosteric binding pocket within the transmembrane domain (Gregory & Conn, 2015). Based on this, sex-dependent changes in splicing, post-translational modifications, or interactions with other proteins (e.g., ER) could disproportionately affect one method of assessing mGlu5 receptor expression. Moving forward, parallel cross-species studies should be designed to use identical techniques to assess potential sex differences in mGlu receptor expression. Ideal studies would examine expression of transcripts, protein, and available receptors for all mGlu receptor subtypes.
Here, we have summarized key pieces of evidence suggesting sex differences in mGlu receptor functions, highlighting literature showing that the gonadal hormone E2 has acute effects on these mGlu receptor functions. However, much less is known about how other factors, including acute effects of other gonadal hormones (e.g., testosterone, progesterone), developmental gonadal hormone effects (i.e., organizational effects of hormones), and sex chromosome complement might influence sex-specific behavioral and neurobiological outcomes. Indeed, there is a rich literature in neuroendocrinology and the sex differences field showing that these other factors influence behavior and neurobiology. For instance, rodent studies have revealed links between circulating testosterone and anxiety-like behaviors. Mice and rats with the testicular feminization mutation, which renders the androgen receptor insensitive to testosterone, exhibit higher anxiety-like behaviors compared to wildtypes (Chen et al., 2014; Zuloaga et al., 2008, 2011). We have also shown that testosterone exposure in adult mice reduces measures of anxiety–/depressive-like behavior after chronic stress exposure in both males and females (Seney et al., 2013a). Whether mGlu receptor signaling mediates the behavioral effects of androgens remains, by and large, an open question.
Additional sources of sex differences may originate in early developmental events and from sex chromosome genes. Exposure to gonadal hormones during sensitive periods of development (“organizational effects”) is known to cause permanent sex differences in behavior and brain structure (Cooke et al., 1998; Handa et al., 1985; Phoenix et al., 1959). Moreover, sex chromosomes differ between males and females in the presence or absence of a Y and in the dosage of X chromosomes. The role of genetic sex has been difficult to investigate due to mosaicism in females due to X-linked inactivation and the relationship between genetic sex and gonadal sex. We and others have begun to parse genetic sex from gonadal sex by using the Four Core Genotypes (FCG) mice. In the FCG strain, the testes determining gene, Sry, was placed on an autosome after spontaneous deletion from the Y chromosome; thus, both XX mice and XY mice can have either ovaries or testes, dissociating genetic from gonadal sex (Arnold & Chen, 2009; McCarthy & Arnold, 2011). Using the FCG mice, we showed that genetic sex influences anxiety-like behavior (Seney et al., 2013a) and gene expression in mood-relevant brain regions (Barko et al., 2019; Paden et al., 2020; Puralewski, Vasilakis, & Seney, 2016; Seney et al., 2013b). Other studies in FCG mice have revealed sex chromosome influences on sex differences in aggressive, parental, social, cognitive, circadian, and substance use-associated behaviors (Aarde et al., 2021; Barker et al., 2010; Cox & Rissman, 2011; Gatewood et al., 2006; Kuljis et al., 2013; Martini et al., 2020; McPhie-Lalmansingh et al., 2008). Together, it is clear that factors other than circulating E2 influence sex-related outcomes, and that there is much work to be done related to how these factors may impact mGlu function.
Acknowledgments
This work was supported in part by the National Institute of Health AA027806 (MEJ) and MH120066 (MLS). CBF was supported by the Center for Neuroscience University of Pittsburgh.
Footnotes
Conflict of Interest
The authors declare no potential conflicts of interest.
References
- Aarde SM, et al. (2021). Sex chromosome complement affects multiple aspects of reversal-learning task performance in mice. Genes, Brain, and Behavior, 20(1), e12685. [DOI] [PubMed] [Google Scholar]
- Abd-Elrahman KS, & Ferguson SSG (2022). Noncanonical metabotropic glutamate receptor 5 signaling in Alzheimer’s disease. Annual Review of Pharmacology and Toxicology, 62, 235–254. [DOI] [PubMed] [Google Scholar]
- Abd-Elrahman KS, et al. (2020). Aβ oligomers induce pathophysiological mGluR5 signaling in Alzheimer’s disease model mice in a sex-selective manner. Science Signaling, 13(662), eabd2494. [DOI] [PubMed] [Google Scholar]
- Akkus F, et al. (2013). Marked global reduction in mGluR5 receptor binding in smokers and ex-smokers determinedby [11C]ABP688 positron emission tomography. Proceedings of the National Academy of Sciences of the United States of America, 110(2), 737–742. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alagarsamy S, Sorensen SD, & Conn PJ (2001). Coordinate regulation of metabotropic glutamate receptors. Current Opinion in Neurobiology, 11(3), 357–362. [DOI] [PubMed] [Google Scholar]
- Altemus M, Sarvaiya N, & Neill Epperson C (2014). Sex differences in anxiety and depression clinical perspectives. Frontiers in Neuroendocrinology, 35(3), 320–330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Angermeyer MC, Goldstein JM, & Kuehn L (1989). Gender differences in schizophrenia: Rehospitalization and community survival. Psychological Medicine, 19(2), 365–382. [DOI] [PubMed] [Google Scholar]
- Angst J, & Dobler-Mikola A (1984). Do the diagnostic criteria determine the sex ratio in depression? Journal of Affective Disorders, 7(3–4), 189–198. [DOI] [PubMed] [Google Scholar]
- Anker JJ, & Carroll ME (2011). Females are more vulnerable to drug abuse than males: Evidence from preclinical studies and the role of ovarian hormones. Current Topics in Behavioral Neurosciences, 8, 73–96. [DOI] [PubMed] [Google Scholar]
- Arnold AP, & Chen X (2009). What does the “four core genotypes” mouse model tell us about sex differences in the brain and other tissues? Frontiers in Neuroendocrinology, 30(1), 1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Backstrom P, et al. (2004). mGluR5 antagonist MPEP reduces ethanol-seeking and relapse behavior. Neuropsychopharmacology, 29(5), 921–928. [DOI] [PubMed] [Google Scholar]
- Ballard TM, et al. (2005). The effect of the mGlu5 receptor antagonist MPEP in rodent tests of anxiety and cognition: A comparison. Psychopharmacology, 179(1), 218–229. [DOI] [PubMed] [Google Scholar]
- Bangasser DA, & Cuarenta A (2021). Sex differences in anxiety and depression: Circuits and mechanisms. Nature Reviews. Neuroscience, 22(11), 674–684. [DOI] [PubMed] [Google Scholar]
- Bara A, et al. (2018). Sex-dependent effects of in utero cannabinoid exposure on cortical function. eLife, 7, e36234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Barker JM, et al. (2010). Dissociation of genetic and hormonal influences on sex differences in alcoholism-related behaviors. The Journal of Neuroscience, 30(27), 9140–9144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Barko K, et al. (2019). Sex-specific effects of stress on mood-related gene expression. Molecular Neuropsychiatry, 5(3), 162–175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Barnes SA, et al. (2015). Disruption of mGluR5 in parvalbumin-positive interneurons induces core features of neurodevelopmental disorders. Molecular Psychiatry, 20(10), 1161–1172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baude A, et al. (1993). The metabotropic glutamate receptor (mGluR 1 alpha) is concentrated at perisynaptic membrane of neuronal subpopulations as detected by immunogold reaction. Neuron, 11(4), 771–787. [DOI] [PubMed] [Google Scholar]
- Belmeguenai A, et al. (2008). Alcohol impairs long-term depression at the cerebellar parallel fiber-Purkinje cell synapse. Journal of Neurophysiology, 100(6), 3167–3174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bender RA, et al. (2017). Sex-dependent regulation of aromatase-mediated synaptic plasticity in the basolateral amygdala. The Journal of Neuroscience, 37(6), 1532–1545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bertaso F, et al. (2010). Homer1a-dependent crosstalk between NMDA and metabotropic glutamate receptors in mouse neurons. PLoS One, 5(3), e9755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Besheer J, et al. (2008). Regulation of motivation to self-administer ethanol by mGluR5 in alcohol-preferring (P) rats. Alcoholism, Clinical and Experimental Research, 32(2), 209–221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bode FJ, et al. (2008). Sex differences in a transgenic rat model of Huntington’s disease: Decreased 17beta-estradiol levels correlate with reduced numbers of DARPP32+ neurons in males. Human Molecular Genetics, 17(17), 2595–2609. [DOI] [PubMed] [Google Scholar]
- Boulware MI, Heisler JD, & Frick KM (2013). The memory-enhancing effects of hippocampal estrogen receptor activation involve metabotropic glutamate receptor signaling. The Journal of Neuroscience, 33(38), 15184–15194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boulware MI, et al. (2005). Estradiol activates group I and II metabotropic glutamate receptor signaling, leading to opposing influences on cAMP response element-binding protein. The Journal of Neuroscience, 25(20), 5066–5078. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bowers ME, & Ressler KJ (2016). Sex-dependence of anxiety-like behavior in cannabinoid receptor 1 (Cnr1) knockout mice. Behavioural Brain Research, 300, 65–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brakeman PR, et al. (1997). Homer: A protein that selectively binds metabotropic glutamate receptors. Nature, 386(6622), 284–288. [DOI] [PubMed] [Google Scholar]
- Busse CS, et al. (2004). The behavioral profile of the potent and selective mGlu5 receptor antagonist 3-[(2-methyl-1,3-thiazol-4-yl)ethynyl]pyridine (MTEP) in rodent models of anxiety. Neuropsychopharmacology, 29(11), 1971–1979. [DOI] [PubMed] [Google Scholar]
- Calipari ES, et al. (2017). Dopaminergic dynamics underlying sex-specific cocaine reward. Nature Communications, 8, 13877. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Campbell RR, et al. (2019). Increased alcohol-drinking induced by manipulations of mGlu5 phosphorylation within the bed nucleus of the Stria terminalis. The Journal of Neuroscience, 39(14), 2745–2761. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cao JK, et al. (2019). Sex-dependent impaired locomotion and motor coordination in the HdhQ200/200 mouse model of Huntington’s disease. Neurobiology of Disease, 132, 104607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carroll ME, et al. (2002). Intravenous cocaine and heroin self-administration in rats selectively bred for differential saccharin intake: Phenotype and sex differences. Psychopharmacology, 161(3), 304–313. [DOI] [PubMed] [Google Scholar]
- Carta M, Mameli M, & Valenzuela CF (2006). Alcohol potently modulates climbing fiber–>Purkinje neuron synapses: Role of metabotropic glutamate receptors. The Journal of Neuroscience, 26(7), 1906–1912. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carzoli KL, et al. (2019). Regulation of NMDA receptor plasticity in the BNST following adolescent alcohol exposure. Frontiers in Cellular Neuroscience, 13, 440. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Casabona G, et al. (1997). Expression and coupling to polyphosphoinositide hydrolysis of group I metabotropic glutamate receptors in early postnatal and adult rat brain. The European Journal of Neuroscience, 9(1), 12–17. [DOI] [PubMed] [Google Scholar]
- Chaban VV, et al. (2003). Estradiol inhibits atp-induced intracellular calcium concentration increase in dorsal root ganglia neurons. Neuroscience, 118(4), 941–948. [DOI] [PubMed] [Google Scholar]
- Chandler LJ, Vaughan DT, & Gass JT (2022). Adolescent alcohol exposure results in sex-specific alterations in conditioned fear learning and memory in adulthood. Frontiers in Pharmacology, 13, 837657. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chávez AE, Chiu CQ, & Castillo PE (2010). TRPV1 activation by endogenous anandamide triggers postsynaptic long-term depression in dentate gyrus. Nature Neuroscience, 13(12), 1511–1518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chavis P, et al. (1994). The metabotropic glutamate receptor types 2/3 inhibit L-type calcium channels via a pertussis toxin-sensitive G-protein in cultured cerebellar granule cells. The Journal of Neuroscience, 14(11 Pt 2), 7067–7076. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen CV, et al. (2014). New knockout model confirms a role for androgen receptors in regulating anxiety-like behaviors and HPA response in mice. Hormones and Behavior, 65(3), 211–218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ciruela F, et al. (2000). Homer-1c/Vesl-1L modulates the cell surface targeting of metabotropic glutamate receptor type 1alpha: Evidence for an anchoring function. Molecular and Cellular Neurosciences, 15(1), 36–50. [DOI] [PubMed] [Google Scholar]
- Conn PJ, & Pin JP (1997). Pharmacology and functions of metabotropic glutamate receptors. Annual Review of Pharmacology and Toxicology, 37, 205–237. [DOI] [PubMed] [Google Scholar]
- Cooke B, et al. (1998). Sexual differentiation of the vertebrate brain: Principles and mechanisms. Frontiers in Neuroendocrinology, 19(4), 323–362. [DOI] [PubMed] [Google Scholar]
- Cox KH, & Rissman EF (2011). Sex differences in juvenile mouse social behavior are influenced by sex chromosomes and social context. Genes, Brain, and Behavior, 10(4), 465–472. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cozzoli DK, et al. (2009). Binge drinking upregulates accumbens mGluR5-Homer2-PI3K signaling: Functional implications for alcoholism. The Journal of Neuroscience, 29(27), 8655–8668. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cozzoli DK, et al. (2014). The effect of mGluR5 antagonism during binge Drinkingon subsequent ethanol intake in C57BL/6J mice: Sex- and age-induced differences. Alcoholism, Clinical and Experimental Research, 38(3), 730–738. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Crews FT, et al. (2016). Adolescent alcohol exposure persistently impacts adult neurobiology and behavior. Pharmacological Reviews, 68(4), 1074–1109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cryan JF, et al. (2003). Antidepressant and anxiolytic-like effects in mice lacking the group III metabotropic glutamate receptor mGluR7. The European Journal of Neuroscience, 17(11), 2409–2417. [DOI] [PubMed] [Google Scholar]
- Davis MJ, et al. (2012). Measures of anxiety, sensorimotor function, and memory in male and female mGluR4−/− mice. Behavioural Brain Research, 229(1), 21–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- De Blasi A, et al. (2001). Molecular determinants of metabotropic glutamate receptor signaling. Trends in Pharmacological Sciences, 22(3), 114–120. [DOI] [PubMed] [Google Scholar]
- De Jesús-Burgos M, Torres-Llenza V, & Pérez-Acevedo NL (2012). Activation of amygdalar metabotropic glutamate receptors modulates anxiety, and risk assessment behaviors in ovariectomized estradiol-treated female rats. Pharmacology, Biochemistry, and Behavior, 101(3), 369–378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- De Jesús-Burgos MI, et al. (2016).Amygdalar activation of group I metabotropic glutamate receptors produces anti- and pro-conflict effects depending upon animal sex in a sexually dimorphic conditioned conflict-based anxiety model. Behavioural Brain Research, 302, 200–212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Di Menna L, et al. (2018). Functional partnership between mGlu3 and mGlu5 metabotropic glutamate receptors in the central nervous system. Neuropharmacology, 128, 301–313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Diehl A, et al. (2007). Alcoholism in women: Is it different in onset and outcome compared to men? European Archives of Psychiatry and Clinical Neuroscience, 257(6), 344–351. [DOI] [PubMed] [Google Scholar]
- Doncheck EM, et al. (2018). 17β-estradiol potentiates the reinstatement of cocaine seeking in female rats: Role of the prelimbic prefrontal cortex and cannabinoid Type-1 receptors. Neuropsychopharmacology, 43(4), 781–790. [DOI] [PMC free article] [PubMed] [Google Scholar]
- DuBois JM, et al. (2016). Characterization of age/sex and the regional distribution of mGluR5 availability in the healthy human brain measured by high-resolution [(11)C] ABP688 PET. European Journal of Nuclear Medicine and Molecular Imaging, 43(1), 152–162. [DOI] [PubMed] [Google Scholar]
- Duvoisin RM, et al. (2010). Sex-dependent cognitive phenotype of mice lacking mGluR8. Behavioural Brain Research, 209(1), 21–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Evans SM, Haney M, & Foltin RW (2002). The effects of smoked cocaine during the follicular and luteal phases of the menstrual cycle in women. Psychopharmacology, 159(4), 397–406. [DOI] [PubMed] [Google Scholar]
- Fagan MP, et al. (2020). Essential and sex-specific effects of mGluR5 in ventromedial hypothalamus regulating estrogen signaling and glucose balance. Proceedings of the National Academy of Sciences of the United States of America, 117(32), 19566–19577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fazio F, et al. (2008). Switch in the expression of mGlu1 and mGlu5 metabotropic glutamate receptors in the cerebellum of mice developing experimental autoimmune encephalomyelitis and in autoptic cerebellar samples from patients with multiple sclerosis. Neuropharmacology, 55(4), 491–499. [DOI] [PubMed] [Google Scholar]
- Feltenstein MW, & See RE (2007). Plasma progesterone levels and cocaine-seeking in freely cycling female rats across the estrous cycle. Drug and Alcohol Dependence, 89(2–3), 183–189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fernandez SM, et al. (2008). Estradiol-induced enhancement of object memory consolidation involves hippocampal extracellular signal-regulated kinase activation and membrane-bound estrogen receptors. The Journal of Neuroscience, 28(35), 8660–8667. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ferretti MT, et al. (2018). Sex differences in Alzheimer disease—The gateway to precision medicine. Nature Reviews. Neurology, 14(8), 457–469. [DOI] [PubMed] [Google Scholar]
- Fisher NM, et al. (2020). Phenotypic profiling of mGlu(7) knockout mice reveals new implications for neurodevelopmental disorders. Genes, Brain, and Behavior, 19(7), e12654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fisher NM, et al. (2021). A GRM7 mutation associated with developmental delay reduces mGlu7 expression and produces neurological phenotypes. JCI Insight, 6(4), e143324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Frank E, Carpenter LL, & Kupfer DJ (1988). Sex differences in recurrent depression: Are there any that are significant? The American Journal of Psychiatry, 145(1), 41–45. [DOI] [PubMed] [Google Scholar]
- Fuentes N, & Silveyra P (2019). Estrogen receptor signaling mechanisms. Advances in Protein Chemistry and Structural Biology, 116, 135–170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- García-Bea A, et al. (2016). Metabotropic glutamate receptor 3 (mGlu3; mGluR3; GRM3) in schizophrenia: Antibody characterisation and a semi-quantitative western blot study. Schizophrenia Research, 177(1–3), 18–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gatewood JD, et al. (2006). Sex chromosome complement and gonadal sex influence aggressive and parental behaviors in mice. The Journal of Neuroscience, 26(8), 2335–2342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gil-Sanz C, et al. (2008). Involvement of the mGluR1 receptor in hippocampal synaptic plasticity and associative learning in behaving mice. Cerebral Cortex, 18(7), 1653–1663. [DOI] [PubMed] [Google Scholar]
- Goddyn H, et al. (2008). Deficits in acquisition and extinction of conditioned responses in mGluR7 knockout mice. Neurobiology of Learning and Memory, 90(1), 103–111. [DOI] [PubMed] [Google Scholar]
- Goldstein JM, & Link BG (1988). Gender and the expression of schizophrenia. Journal of Psychiatric Research, 22(2), 141–155. [DOI] [PubMed] [Google Scholar]
- Goldstein JM, Tsuang MT, & Faraone SV (1989). Gender and schizophrenia: Implications for understanding the heterogeneity of the illness. Psychiatry Research, 28(3), 243–253. [DOI] [PubMed] [Google Scholar]
- Goldstein JM, et al. (1998). Are there sex differences in neuropsychological functions among patients with schizophrenia? The American Journal of Psychiatry, 155(10), 1358–1364. [DOI] [PubMed] [Google Scholar]
- Gregory KJ, & Conn PJ (2015). Molecular insights into metabotropic glutamate receptor allosteric modulation. Molecular Pharmacology, 88(1), 188–202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gross WC, & Billingham RE (1998). Alcohol consumption and sexual victimization among college women. Psychological Reports, 82(1), 80–82. [DOI] [PubMed] [Google Scholar]
- Gross KS, et al. (2016). Opposite effects of mGluR1a and mGluR5 activation on nucleus Accumbens medium spiny neuron dendritic spine density. PLoS One, 11(9), e0162755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gross KS, et al. (2018). mGluR5mediates dihydrotestosterone-induced nucleus Accumbens structural plasticity, but not conditioned reward. Frontiers in Neuroscience, 12, 855. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grove-Strawser D, Boulware MI, & Mermelstein PG (2010). Membrane estrogen receptors activate the metabotropic glutamate receptors mGluR5 and mGluR3 to bidirectionally regulate CREB phosphorylation in female rat striatal neurons. Neuroscience, 170(4), 1045–1055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grueter BA, Brasnjo G, & Malenka RC (2010). Postsynaptic TRPV1 triggers cell type-specific long-term depression in the nucleus accumbens. Nature Neuroscience, 13(12), 1519–1525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Haas LT, et al. (2017). Silent allosteric modulation of mGluR5 maintains glutamate signaling while rescuing Alzheimer’s mouse phenotypes. Cell Reports, 20(1), 76–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hafner H, et al. (1989). How does gender influence age at first hospitalization for schizophrenia? A transnational case register study. Psychological Medicine, 19(4), 903–918. [DOI] [PubMed] [Google Scholar]
- Hambrecht M, Maurer K, & Hafner H (1992). Gender differences in schizophrenia in three cultures. Results of the WHO collaborative study on psychiatric disability. Social Psychiatry and Psychiatric Epidemiology, 27(3), 117–121. [DOI] [PubMed] [Google Scholar]
- Hamilton JA, Grant M, & Jensvold MF (1996). Sex and treatment of depression: When does it matter? In Jensvold MF, Halbreich U, & Hamilton JA (Eds.), Psychopharmacology and women: Sex, gender, and hormones (pp. 241–257). Washington, DC: American Psychiatric Press. [Google Scholar]
- Handa RJ, et al. (1985). Differential effects of the perinatal steroid environment on three sexually dimorphic parameters of the rat brain. Biology of Reproduction, 32(4), 855–864. [DOI] [PubMed] [Google Scholar]
- Hartmann J, Henning HA, & Konnerth A (2011). mGluR1/TRPC3-mediated synaptic transmission and calcium signaling in mammalian central neurons. Cold Spring Harbor Perspectives in Biology, 3(4), a006726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hermans E, & Challiss RA (2001). Structural, signalling and regulatory properties of the group I metabotropic glutamate receptors: Prototypic family C G-protein-coupled receptors. The Biochemical Journal, 359(Pt 3), 465–484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Highland JN, et al. (2019). Group II metabotropic glutamate receptor blockade promotes stress resilience in mice. Neuropsychopharmacology, 44(10), 1788–1796. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hill MN, Karacabeyli ES, & Gorzalka BB (2007). Estrogen recruits the endocannabinoid system to modulate emotionality. Psychoneuroendocrinology, 32(4), 350–357. [DOI] [PubMed] [Google Scholar]
- Hodge CW, et al. (2006). The mGluR5 antagonist MPEP selectively inhibits the onset and maintenance of ethanol self-administration in C57BL/6J mice. Psychopharmacology, 183(4), 429–438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hölscher C, et al. (2004). Lack of the metabotropic glutamate receptor subtype 7 selectively impairs short-term working memory but not long-term memory. Behavioural Brain Research, 154(2), 473–481. [DOI] [PubMed] [Google Scholar]
- Hu M, & Becker JB (2003). Effects of sex and estrogen on behavioral sensitization to cocaine in rats. The Journal of Neuroscience, 23(2), 693–699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang G, Thompson SL, & Taylor JR (2021). MPEP lowers binge drinking in male and female C57BL/6 mice: Relationship with mGlu5/Homer2/Erk2 signaling. Alcoholism, Clinical and Experimental Research, 45(4), 732–742. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang GZ, & Woolley CS (2012). Estradiol acutely suppresses inhibition in the hippocampus through a sex-specific endocannabinoid and mGluR-dependent mechanism. Neuron, 74(5), 801–808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huber G, et al. (1980). Longitudinal studies of schizophrenic patients. Schizophrenia Bulletin, 6(4), 592–605. [DOI] [PubMed] [Google Scholar]
- Jackson LR, Robinson TE, & Becker JB (2006). Sex differences and hormonal influences on acquisition of cocaine self-administration in rats. Neuropsychopharmacology, 31(1), 129–138. [DOI] [PubMed] [Google Scholar]
- Joffe ME, et al. (2018). Metabotropic glutamate receptors in alcohol use disorder: Physiology, plasticity, and promising pharmacotherapies. ACS Chemical Neuroscience, 9(9), 2188–2204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Joffe ME, et al. (2019). Metabotropic glutamate receptor subtype 3 gates acute stress-induced dysregulation of amygdalo-cortical function. Molecular Psychiatry, 24(6), 916–927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Joffe ME, et al. (2020). mGlu2 and mGlu3 negative allosteric modulators divergently enhance Thalamocortical transmission and exert rapid antidepressant-like effects. Neuron, 105(1), 46–59.e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Joffe ME, et al. (2021). Frontal cortex genetic ablation of metabotropic glutamate receptor subtype 3 (mGlu(3)) impairs postsynaptic plasticity and modulates affective behaviors. Neuropsychopharmacology, 46(12), 2148–2157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Joffe M, et al. (2022). Acute restraint stress redirects prefrontal cortex circuit function through mGlu5 receptor plasticity on somatostatin-expressing interneurons. Neuron, 110(6), 1068–1083.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Johnson AR, et al. (2019). Cues play a critical role in estrous cycle-dependent enhancement of cocaine reinforcement. Neuropsychopharmacology, 44(7), 1189–1197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jong YJ, et al. (2005). Functional metabotropic glutamate receptors on nuclei from brain and primary cultured striatal neurons. Role of transporters in delivering ligand. The Journal of Biological Chemistry, 280(34), 30469–30480. [DOI] [PubMed] [Google Scholar]
- Kalinichev M, et al. (2014). Characterization of the novel positive allosteric modulator of the metabotropic glutamate receptor 4 ADX88178 in rodent models of neuropsychiatric disorders. The Journal of Pharmacology and Experimental Therapeutics, 350(3), 495–505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kammermeier PJ, et al. (2000). Homer proteins regulate coupling of group I metabotropic glutamate receptors to N-type calcium and M-type potassium channels. The Journal of Neuroscience, 20(19), 7238–7245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kasten CR, et al. (2020). Adolescent alcohol exposure produces sex differences in negative affect-like behavior and group I mGluR BNST plasticity. Neuropsychopharmacology, 45(8), 1306–1315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kasten CR, et al. (2021). BNST specific mGlu5 receptor knockdown regulates sex-dependent expression of negative affect produced by adolescent ethanol exposure and adult stress. Translational Psychiatry, 11(1), 178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kessler RC, et al. (2005). Lifetime prevalence and age-of-onset distributions of DSM-IV disorders in the National Comorbidity Survey Replication. Archives of General Psychiatry, 62(6), 593–602. [DOI] [PubMed] [Google Scholar]
- Kessler RC, et al. (2012). Twelve-month and lifetime prevalence and lifetime morbid risk of anxiety and mood disorders in the United States. International Journal of Methods in Psychiatric Research, 21(3), 169–184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kornstein SG, et al. (2000a). Gender differences in chronic major and double depression. Journal of Affective Disorders, 60(1), 1–11. [DOI] [PubMed] [Google Scholar]
- Kornstein SG, et al. (2000b). Gender differences in treatment response to sertraline versus imipramine in chronic depression. The American Journal of Psychiatry, 157(9), 1445–1452. [DOI] [PubMed] [Google Scholar]
- Kuljis DA, et al. (2013). Gonadal- and sex-chromosome-dependent sex differences in the circadian system. Endocrinology, 154(4), 1501–1512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lebow MA, & Chen A (2016). Overshadowed by the amygdala: The bed nucleus of the stria terminalis emerges as key to psychiatric disorders. Molecular Psychiatry, 21(4), 450–463. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Levin ER (2009). Plasma membrane estrogen receptors. Trends in Endocrinology and Metabolism, 20(10), 477–482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lewis B, Hoffman LA, & Nixon SJ (2014). Sex differences in drug use among polysubstance users. Drug and Alcohol Dependence, 145, 127–133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li SH, et al. (2022). Metabotropic glutamate receptor 5 antagonism reduces pathology and differentially improves symptoms in male and female heterozygous zQ175 Huntington’s mice. Frontiers in Molecular Neuroscience, 15, 801757. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Logan CN, et al. (2020). Ceftriaxone and mGlu2/3 interactions in the nucleus accumbens core affect the reinstatement of cocaine-seeking in male and female rats. Psychopharmacology, 237(7), 2007–2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lominac KD, et al. (2006). Behavioral and neurochemical interactions between group 1 mGluR antagonists and ethanol: Potential insight into their anti-addictive properties. Drug and Alcohol Dependence, 85(2), 142–156. [DOI] [PubMed] [Google Scholar]
- Loweth JA, et al. (2014). Synaptic depression via mGluR1 positive allosteric modulation suppresses cue-induced cocaine craving. Nature Neuroscience, 17(1), 73–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Luscher C, & Huber KM (2010). Group 1 mGluR-dependent synaptic long-term depression: Mechanisms and implications for circuitry and disease. Neuron, 65(4), 445–459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lynch WJ, et al. (2001). Role of estrogen in the acquisition of intravenously self-administered cocaine in female rats. Pharmacology, Biochemistry, and Behavior, 68(4), 641–646. [DOI] [PubMed] [Google Scholar]
- Maksymetz J, & Joffe ME (2021). mGlu Receptor modulation in murine models of stress and affective disorders. In Olive MF, Burrows BT, & Leyrer-Jackson JM (Eds.), Metabotropic glutamate receptor technologies (pp. 259–296). New York, NY: Humana. [Google Scholar]
- Mao LM, & Wang JQ (2016). Regulation of group I metabotropic glutamate receptors by MAPK/ERK in neurons. Journal of Nature and Science, 2(12), e268. [PMC free article] [PubMed] [Google Scholar]
- Martin LJ, et al. (1992). Cellular localization of a metabotropic glutamate receptor in rat brain. Neuron, 9(2), 259–270. [DOI] [PubMed] [Google Scholar]
- Martinez LA, et al. (2014). Estradiol facilitation of cocaine-induced locomotor sensitization in female rats requires activation of mGluR5. Behavioural Brain Research, 271, 39–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Martini M, et al. (2020). Sex chromosome complement influences vulnerability to cocaine in mice. Hormones and Behavior, 125, 104821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Matas E, et al. (2021). Major motor and gait deficits with sexual dimorphism in a Shank3 mutant mouse model. Molecular Autism, 12(1), 2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mathews DC, Henter ID, & Zarate CA (2012). Targeting the glutamatergic system to treat major depressive disorder: Rationale and progress to date. Drugs, 72(10), 1313–1333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McCarthy MM, & Arnold AP (2011). Reframing sexual differentiation of the brain. Nature Neuroscience, 14(6), 677–683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McEwen B, et al. (2001). Tracking the estrogen receptor in neurons: Implications for estrogen-induced synapse formation. Proceedings of the National Academy of Sciences of the United States of America, 98(13), 7093–7100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McPhie-Lalmansingh AA, et al. (2008). Sex chromosome complement affects social interactions in mice. Hormones and Behavior, 54(4), 565–570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Meitzen J, & Mermelstein PG (2011). Estrogen receptors stimulate brain region specific metabotropic glutamate receptors to rapidly initiate signal transduction pathways. Journal of Chemical Neuroanatomy, 42(4), 236–241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Meitzen J, et al. (2019). The expression of select genes necessary for membrane-associated estrogen receptor signaling differ by sex in adult rat hippocampus. Steroids, 142, 21–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mercer AA, et al. (2016). Sex differences in cerebellar synaptic transmission and sex-specific responses to autism-linked Gabrb3 mutations in mice. eLife, 5, e07596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mermelstein PG, Becker JB, & Surmeier DJ (1996). Estradiol reduces calcium currents in rat neostriatal neurons via a membrane receptor. The Journal of Neuroscience, 16(2), 595–604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Milner TA, et al. (2001). Ultrastructural evidence that hippocampal alpha estrogen receptors are located at extranuclear sites. The Journal of Comparative Neurology, 429(3), 355–371. [PubMed] [Google Scholar]
- Minami K, et al. (1998). Effects of ethanol and anesthetics on type 1 and 5 metabotropic glutamate receptors expressed in Xenopus laevis oocytes. Molecular Pharmacology, 53(1), 148–156. [DOI] [PubMed] [Google Scholar]
- Mitterling KL, et al. (2010). Cellular and subcellular localization of estrogen and progestin receptor immunoreactivities in the mouse hippocampus. The Journal of Comparative Neurology, 518(14), 2729–2743. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mora S, Dussaubat N, & Díaz-Véliz G (1996). Effects of the estrous cycle and ovarian hormones on behavioral indices of anxiety in female rats. Psychoneuroendocrinology, 21(7), 609–620. [DOI] [PubMed] [Google Scholar]
- Murphy MP, & LeVine H 3rd. (2010). Alzheimer’s disease and the amyloid-beta peptide. Journal of Alzheimer’s Disease, 19(1), 311–323. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Najt P, Fusar-Poli P, & Brambilla P (2011). Co-occurring mental and substance abuse disorders: A review on the potential predictors and clinical outcomes. Psychiatry Research, 186(2–3), 159–164. [DOI] [PubMed] [Google Scholar]
- Nardecchia F, et al. (2018). Targeting mGlu5 metabotropic glutamate receptors in the treatment of cognitive dysfunction in a mouse model of phenylketonuria. Frontiers in Neuroscience, 12, 154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Netzeband JG, & Gruol DL (1995). Modulatory effects of acute ethanol on metabotropic glutamate responses in cultured Purkinje neurons. Brain Research, 688(1–2), 105–113. [DOI] [PubMed] [Google Scholar]
- Ning LN, et al. (2018). Gender-related hippocampal proteomics study from Young rats after chronic unpredicted mild stress exposure. Molecular Neurobiology, 55(1), 835–850. [DOI] [PubMed] [Google Scholar]
- Niswender CM, & Conn PJ (2010). Metabotropic glutamate receptors: Physiology, pharmacology, and disease. Annual Review of Pharmacology and Toxicology, 50, 295–322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nixon SJ, Tivis R, & Parsons OA (1995). Behavioral dysfunction and cognitive efficiency in male and female alcoholics. Alcoholism, Clinical and Experimental Research, 19(3), 577–581. [DOI] [PubMed] [Google Scholar]
- Oberlander JG, & Woolley CS (2016). 17β-estradiol acutely potentiates glutamatergic synaptic transmission in the Hippocampus through distinct mechanisms in males and females. The Journal of Neuroscience, 36(9), 2677–2690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ohno-Shosaku T, et al. (2002). Cooperative endocannabinoid production by neuronal depolarization and group I metabotropic glutamate receptor activation. The European Journal of Neuroscience, 15(6), 953–961. [DOI] [PubMed] [Google Scholar]
- Paden W, et al. (2020). Sex differences in adult mood and in stress-induced transcriptional coherence across mesocorticolimbic circuitry. Translational Psychiatry, 10(1), 59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Palucha A, & Pilc A (2007). Metabotropic glutamate receptor ligands as possible anxiolytic and antidepressant drugs. Pharmacology & Therapeutics, 115(1), 116–147. [DOI] [PubMed] [Google Scholar]
- Peixoto RT, et al. (2016). Early hyperactivity and precocious maturation of corticostriatal circuits in Shank3B(−/−) mice. Nature Neuroscience, 19(5), 716–724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Peltier MR, et al. (2019). Sex differences in stress-related alcohol use. Neurobiology of Stress, 10, 100149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Peterson BM, Mermelstein PG, & Meisel RL (2015). Estradiol mediates dendritic spine plasticity in the nucleus accumbens core through activation of mGluR5. Brain Structure & Function, 220(4), 2415–2422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Phoenix CH, et al. (1959). Organizing action of prenatally administered testosterone propionate on the tissues mediating mating behavior in the female guinea pig. Endocrinology, 65, 369–382. [DOI] [PubMed] [Google Scholar]
- Pietraszek M, et al. (2005).Anxiolytic-like effects of mGlu1 and mGlu5 receptor antagonists in rats. European Journal of Pharmacology, 514(1), 25–34. [DOI] [PubMed] [Google Scholar]
- Puralewski R, Vasilakis G, & Seney ML (2016). Sex-related factors influence expression of mood-related genes in the basolateral amygdala differentially depending on age and stress exposure. Biology of Sex Differences, 7, 50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Purgert CA, et al. (2014). Intracellular mGluR5 can mediate synaptic plasticity in the hippocampus. The Journal of Neuroscience, 34(13), 4589–4598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Quitkin FM, et al. (2002). Are there differences between women’s and men’s antidepressant responses? The American Journal of Psychiatry, 159(11), 1848–1854. [DOI] [PubMed] [Google Scholar]
- Randall CL, et al. (1999). Telescoping of landmark events associated with drinking: A gender comparison. Journal of Studies on Alcohol, 60(2), 252–260. [DOI] [PubMed] [Google Scholar]
- Razandi M, et al. (2004). Plasma membrane estrogen receptors exist and functions as dimers. Molecular Endocrinology, 18(12), 2854–2865. [DOI] [PubMed] [Google Scholar]
- Renner M, et al. (2010). Deleterious effects of amyloid beta oligomers acting as an extracellular scaffold for mGluR5. Neuron, 66(5), 739–754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ribeiro FM, et al. (2014). Metabotropic glutamate receptor 5 knockout promotes motor and biochemical alterations in a mouse model of Huntington’s disease. Human Molecular Genetics, 23(8), 2030–2042. [DOI] [PubMed] [Google Scholar]
- Roberts DC, Bennett SA, & Vickers GJ (1989). The estrous cycle affects cocaine self-administration on a progressive ratio schedule in rats. Psychopharmacology, 98(3), 408–411. [DOI] [PubMed] [Google Scholar]
- Roche KW, et al. (1999). Homer 1b regulates the trafficking of group I metabotropic glutamate receptors. The Journal of Biological Chemistry, 274(36), 25953–25957. [DOI] [PubMed] [Google Scholar]
- Romano C, et al. (1995). Distribution of metabotropic glutamate receptor mGluR5 immunoreactivity in rat brain. The Journal of Comparative Neurology, 355(3), 455–469. [DOI] [PubMed] [Google Scholar]
- Salokangas RK (1983). Prognostic implications of the sex of schizophrenic patients. The British Journal of Psychiatry, 142, 145–151. [DOI] [PubMed] [Google Scholar]
- Sartorius N, et al. (1986). Early manifestations and first-contact incidence of schizophrenia in different cultures. A preliminary report on the initial evaluation phase of the WHO collaborative study on determinants of outcome of severe mental disorders. Psychological Medicine, 16(4), 909–928. [DOI] [PubMed] [Google Scholar]
- Schiefer J, et al. (2004). The metabotropic glutamate receptor 5 antagonist MPEP and the mGluR2 agonist LY379268 modify disease progression in a transgenic mouse model of Huntington’s disease. Brain Research, 1019(1–2), 246–254. [DOI] [PubMed] [Google Scholar]
- Seney ML, et al. (2013a). The role of genetic sex in affect regulation and expression of GABA-related genes across species. Frontiers in Psychiatry, 4, 104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Seney ML, et al. (2013b). Sex chromosome complement regulates expression of mood-related genes. Biology of Sex Differences, 4(1), 20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Silverstein B. (1999). Gender difference in the prevalence of clinical depression: The role played by depression associated with somatic symptoms. The American Journal of Psychiatry, 156(3), 480–482. [DOI] [PubMed] [Google Scholar]
- Simonyi A, Schachtman TR, & Christoffersen GR (2005). The role of metabotropic glutamate receptor 5 in learning and memory processes. Drug News & Perspectives, 18(6), 353–361. [DOI] [PubMed] [Google Scholar]
- Smart K, et al. (2019). Sex differences in [(11)C]ABP688 binding: A positron emission tomography study of mGlu5 receptors. European Journal of Nuclear Medicine and Molecular Imaging, 46(5), 1179–1183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Szumlinski KK, Ary AW, & Lominac KD (2008). Homers regulate drug-induced neuroplasticity: Implications for addiction. Biochemical Pharmacology, 75(1), 112–133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Szumlinski KK, et al. (2004). Homer proteins regulate sensitivity to cocaine. Neuron, 43(3), 401–413. [DOI] [PubMed] [Google Scholar]
- Szumlinski KK, et al. (2005). Homer2 is necessary for EtOH-induced neuroplasticity. The Journal of Neuroscience, 25(30) 7054–7061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Szumlinski KK, et al. (2008). Accumbens Homer2 overexpression facilitates alcohol-induced neuroplasticity in C57BL/6J mice. Neuropsychopharmacology, 33(6), 1365–1378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tabatadze N, et al. (2015). Sex differences in molecular signaling at inhibitory synapses in the Hippocampus. The Journal of Neuroscience, 35(32), 11252–11265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tonn Eisinger KR, et al. (2018). Interactions between estrogen receptors and metabotropic glutamate receptors and their impact on drug addiction in females. Hormones and Behavior, 104, 130–137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Torres ERS, et al. (2018). Effects of sub-chronic MPTP exposure on behavioral and cognitive performance and the microbiome of wild-type and mGlu8 knockout female and male mice. Frontiers in Behavioral Neuroscience, 12, 140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tu JC, et al. (1999). Coupling of mGluR/Homer and PSD-95 complexes by the shank family of postsynaptic density proteins. Neuron, 23(3), 583–592. [DOI] [PubMed] [Google Scholar]
- Tuyns AJ, & Pequignot G (1984). Greater risk of ascitic cirrhosis in females in relation to alcohol consumption. International Journal of Epidemiology, 13(1), 53–57. [DOI] [PubMed] [Google Scholar]
- Uchino S, & Waga C (2013). SHANK3 as an autism spectrum disorder-associated gene. Brain & Development, 35(2), 106–110. [DOI] [PubMed] [Google Scholar]
- Urbano-Marquez A, et al. (1995). The greater risk of alcoholic cardiomyopathy and myopathy in women compared with men. JAMA, 274(2), 149–154. [DOI] [PubMed] [Google Scholar]
- Valenti O, Conn PJ, & Marino MJ (2002). Distinct physiological roles of the Gq-coupled metabotropic glutamate receptors co-expressed in the same neuronal populations. Journal of Cellular Physiology, 191(2), 125–137. [DOI] [PubMed] [Google Scholar]
- Van den Pol AN (1994). Metabotropic glutamate receptor mGluR1 distribution and ultrastructural localization in hypothalamus. The Journal of Comparative Neurology, 349(4), 615–632. [DOI] [PubMed] [Google Scholar]
- van den Pol AN, Romano C, & Ghosh P (1995). Metabotropic glutamate receptor mGluR5 subcellular distribution and developmental expression in hypothalamus. The Journal of Comparative Neurology, 362(1), 134–150. [DOI] [PubMed] [Google Scholar]
- Varma N, et al. (2001). Metabotropic glutamate receptors drive the endocannabinoid system in hippocampus. The Journal of Neuroscience, 21(24), Rc188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Verpelli C, et al. (2011). Importance of Shank3 protein in regulating metabotropic glutamate receptor 5 (mGluR5) expression and signaling at synapses. The Journal of Biological Chemistry, 286(40), 34839–34850. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang X, et al. (2011). Synaptic dysfunction and abnormal behaviors in mice lacking major isoforms of Shank3. Human Molecular Genetics, 20(15), 3093–3108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang Y, et al. (2015). Prenatal chronic mild stress induces depression-like behavior and sex-specific changes in regional glutamate receptor expression patterns in adult rats. Neuroscience, 301, 363–374. [DOI] [PubMed] [Google Scholar]
- Watson CS, Jeng YJ, & Kochukov MY (2008). Nongenomic actions of estradiol compared with estrone and estriol in pituitary tumor cell signaling and proliferation. The FASEB Journal, 22(9), 3328–3336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weiland NG, et al. (1997). Distribution and hormone regulation of estrogen receptor immunoreactive cells in the hippocampus of male and female rats. The Journal of Comparative Neurology, 388(4), 603–612. [DOI] [PubMed] [Google Scholar]
- Wickens MM, Bangasser DA, & Briand LA (2018). Sex differences in psychiatric disease: A focus on the glutamate system. Frontiers in Molecular Neuroscience, 11, 197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Woolley CS, & McEwen BS (1993). Roles of estradiol and progesterone in regulation of hippocampal dendritic spine density during the estrous cycle in the rat. The Journal of Comparative Neurology, 336(2), 293–306. [DOI] [PubMed] [Google Scholar]
- Woolley CS, et al. (1990). Naturally occurring fluctuation in dendritic spine density on adult hippocampal pyramidal neurons. The Journal of Neuroscience, 10(12), 4035–4039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xiao MY, Gustafsson B, & Niu YP (2006). Metabotropic glutamate receptors in the trafficking of ionotropic glutamate and GABA(A) receptors at central synapses. Current Neuropharmacology, 4(1), 77–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xiao B, et al. (1998). Homer regulates the association of group 1 metabotropic glutamate receptors with multivalent complexes of homer-related, synaptic proteins. Neuron, 21(4), 707–716. [DOI] [PubMed] [Google Scholar]
- Xie X, et al. (2010). Effects of mGluR1 antagonism in the dorsal hippocampus on drug context-induced reinstatement of cocaine-seeking behavior in rats. Psychopharmacology, 208(1), 1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu J, et al. (2009). mGluR5 has a critical role in inhibitory learning. The Journal of Neuroscience, 29(12), 3676–3684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Young MA, et al. (1990). Sex differences in the lifetime prevalence of depression: Does varying the diagnostic criteria reduce the female/male ratio? Journal of Affective Disorders, 18(3), 187–192. [DOI] [PubMed] [Google Scholar]
- Yu F, et al. (2013). Metabotropic glutamate receptor I (mGluR1) antagonism impairs cocaine-induced conditioned place preference via inhibition of protein synthesis. Neuropsychopharmacology, 38(7), 1308–1321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhao Z, Fan L, & Frick KM (2010). Epigenetic alterations regulate estradiol-induced enhancement of memory consolidation. Proceedings of the National Academy of Sciences of the United States of America, 107(12), 5605–5610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhao Z, et al. (2012). Hippocampal histone acetylation regulates object recognition and the estradiol-induced enhancement of object recognition. The Journal of Neuroscience, 32(7), 2344–2351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zielonka D, et al. (2013). The influence of gender on phenotype and disease progression in patients with Huntington’s disease. Parkinsonism & Related Disorders, 19(2), 192–197. [DOI] [PubMed] [Google Scholar]
- Zoicas I, & Kornhuber J (2019). The role of metabotropic glutamate receptors in social behavior in rodents. International Journal of Molecular Sciences, 20(6), 1412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zuloaga DG, et al. (2008). Mice with the testicular feminization mutation demonstrate a role for androgen receptors in the regulation of anxiety-related behaviors and the hypothalamic-pituitary-adrenal axis. Hormones and Behavior, 54(5), 758–766. [DOI] [PubMed] [Google Scholar]
- Zuloaga DG, et al. (2011). Male rats with the testicular feminization mutation of the androgen receptor display elevated anxiety-related behavior and corticosterone response to mild stress. Hormones and Behavior, 60(4), 380–388. [DOI] [PMC free article] [PubMed] [Google Scholar]