Supplementary Material: Converging roles of glutamate receptors in domestication and prosociality

Thomas O’Rourke1,2 and Cedric Boeckx1,2,3 1Universitat de Barcelona 2Universitat de Barcelona Institute of Complex Systems 3ICREA

October 10, 2018

S1 Broader changes to glutamatergic signaling under domestication

Signals of selection have been detected in AMH and Yaks on NETO2 [1,2], which encodes an accessory subunit that modulates the desensitization of kainate receptors, and interacts with the GRIP scaffolding to increase receptor abundance at synapses [3,4]. In mouse hippocampal cultures, NETO2 and GRIK2 interact to increase recycling of the cotransporter SLC12A5 to the surface membrane [5] (signals of selection on the corresponding in AMH [6]). Increased abundance of SLC12A5 at hippocampal dendritic spines suppresses AMPA mediated excitatory transmission by preventing the trans- membrane diffusion of GRIA1 proteins [7], and NETO2-SLC12A5 interactions during development promote increases in GABAergic inhibitory signaling in the [8]. Altered prefrontal expression of different SLC12A5 tran- scripts have been associated with schizophrenia and affective disorders [9], and this gene has also been associated with autism, epilepsy and developmental delay [10, 11, 12]. The signals of selection on NETO2 and SLC12A5, their postsynap- tic interactions with kainate receptors, regulation of excitatory and inhibitory transmission, and associations with affective and developmental disorders make them candidate interactors for the modulation of the stress response in both domesticate species and humans. In the case of rats selected for tame behaviors, expression differences are also prominent in glutamatergic-signaling implicated in the stress response. Most notably, SLC17A7, which encodes for a vesicular glutamate transporter, is expressed twice as abundantly in the brains of tame rats as in those of the

1 aggressive strain. Underexpression of SLC17A7 has been implicated in increased anxiety in mice [13]. The postsynaptic scaffolding proteins DLGAP1 and DLGAP3, have both been implicated in OCD and Tourette’s Syndrome [6,1, 14, 15, 16]. DLGAP1 and GRIK2 have been identified in a genome-wide association study as high- confidence interactors in the etiology of OCD [17]. Signals of selection have been identified on DLGAP1 in a horse domestication study [18]. Stereotyped and compulsive behaviors are common in horses, as well as many other domes- ticated animals [19, 20]. Indeed, pharmacological targeting of glutamatergic signaling is successful in treating both Canine Compulsive Disorder and human OCD, suggesting a shared etiology for these conditions [21]. Both DLGAP1 and DLGAP3 (signals of selection in AMH [1,6]) have been implicated in Tourette’s syndrome and OCD, as well as schizophrenia and major depression. At the , DLGAP proteins interact with DLG membrane- associated proteins, and associate with kainate receptors. Signals of selection have been detected in on DLG4 in yaks [2]. When cotransfected in cell cul- tures, GRIK2 subunits associate with DLGAP3 only when accompanied by DLG4 [22]. GRIK2/GRIK5 heteromeric receptors desensitize more slowly to glutamate when they associate with DLG4 [23]. DLGAP1 knockout mice exhibit marked social deficits, correlating with loss of associations between postsynaptic scaffolding complexes and the DLG membrane-associated proteins [24]. DLGAP3 knockout mice exhibit OCD like behaviors, and have increased NMDA and reduced AMPA transmission at corti- costriatal synapses [25]. Knockout of all five genes (GRIK1-5 ) results in comparable preservative behaviors, yet also leads to reduced NMDA: AMPA ratios at corticostriatal synapses [22]. The same effects were not found in CA3 of the hippocampus, suggesting that kainate receptors are not essen- tial for NMDA mobilization in this region. Postsynaptically localized GRIK2 receptors inhibit glutamate signaling in striatal direct pathway medium spiny by activating presynaptic cannabinoid CB1 (G-protein-coupled) recep- tors, thus regulating plasticity in basal ganglia circuits [26]. The interactions of kainate receptors with DLG and DLGAP proteins, their roles in the reg- ulation of striatal plasticity, and their involvement overlapping stress-related phenotypes make them plausible candidates for having come under convergent selection in domestication and modern human evolution. Many glutamate signaling genes have been identified in a recent study of genomic regions associated with tame behaviors in the domesticated fox [27]. These include SORCS1, whose encoded protein is involved in trafficking and maintaining AMPA receptors at the cell membrane, increasing synaptic trans- mission [28]. This gene was detected alongside other glutamatergic signaling genes, including SORCS2, SORCS3 (identified in the fox study), and the AMPA receptor gene GRIA1 in a draft human selective sweep study [29]. However, in a revised version of the same paper, only signals on SORCS2 remained [6]. The SORCS2 protein is highly enriched on post-synaptic densities in the stria- tum and hippocampus, where it regulates NMDA-dependent and spatial learning [30]. SORCS2 knockout mice display manic-depressive

2 symptoms, and polymorphisms in the human population have been associated with bipolar disorder, schizophrenia, and attention deficit hyperactivity disorder (ADHD) [31]. SORCS3 knockout mice have fear-extinction and spatial- deficits [32]. Each of the above SORCS proteins interact with DLG4 [28, 30, 32]. PLCB1 and PLCB4 display signals of selection in red foxes, pigs, and yaks [27, 33,2]. PLCB4 acts downstream of GRM1 synaptic signaling to mobilize endocannabinoids, which suppress glutamate release [34]. In humans, this gene has been implicated in syndromic malformations of the ear and jaw [35]. PLCB1 also modulates GRM signaling and has been implicated in schizophrenia [36, 37]. Signals on KCNJ3 have been identified in foxes and multiple human selective sweep studies [1, 29, 14, 27]. This gene is expressed in the adrenal gland, and its encoded protein has been implicated in cell guidance and localization during CNS development, associates with postsynaptic glutamatergic proteins, and has been implicated in schizophrenia [38, 39, 40]. CACNA1C, which encodes for a voltage-dependent calcium channel subunit, has been detected in a cattle domestication study, and distinct allelic variants are associated with tame and aggressive breeds of foxes [41, 27]. In humans, this gene has been associated with bipolar disorder, schizophrenia, and major depression [42]. A single polymorphism on CACNA1C in schizophrenics is associated with increased expression and higher activity in the hippocampus in response to emotional stimuli [43], as well as higher left amygdala activity in bipolar disorder [44]. CACNA1D has been detected in human and horse selective sweep studies [1, 18]. Both CACNA1C and CACNA1D are abundantly expressed in striatal medium spiny neurons (MSNs) [45]. CACNA1D subunits bind to glutamatergic scaffolding proteins of striatopallidal MSNs, where they inhibit glutamate transmission by allowing Ca2+ influx, thus downregulating the dendritic spine densities. MSNs are targeted by dopaminergic afferents which drive glutamatergic transmission and inhibit CACNA1D channel opening. When dopaminergic signaling is abolished, spine densities are decreased via the opening of CACNA1D channels in a process thought to be critical in the motor deficits experienced by Parkinson’s disease patients [46]. CACNA1D is also expressed in the adrenal gland [47]. The abundant striatal expression of kainate and other glutamatergic signal- ing genes, their interactions at synapses, overlapping involvement in cognitive and stress disorders, including OCD, and associations with tame behaviors in the fox, suggest that convergent selection on striatal-mediated behaviors may have accompanied modulation of the HPA stress response. In the case of hu- mans, it has recently been argued that increases in prosociality following our divergence from other extant primate lineages was driven by neurochemical changes, especially to dopaminergic signaling, in the striatum [48]. We propose that such selective pressures on prosociality have continued following our split from Neanderthals, and that convergent signals of selection with domesticates across the glutamatergic system are indicative of this. SLITRK1, which encodes for a transmembrane protein at excitatory synapses, has a high-frequency missense change in modern humans and may be under positive selection in our lineage [49,6, 50]. This gene has also been impli-

3 cated in OCD and Tourette’s syndrome [51]. Beyond the receptor genes high- lighted in the present study, OCD is associated with a number of genes related to glutamatergic signaling that show signals of selection in AMH. These in- clude SLC1A1, which encodes for a neuronal glutamate transporter, is expressed prominently on hippocampal dendrites, and is most consistently associated with OCD among glutamatergic signaling genes [6, 52, 53, 54].

S2 Kainate and metabotropic receptor genes in human and domesticated behavioral pheno- types Kainate receptor genes in human and domesticated behav- ioral phenotypes GRIK3 has been implicated in artificially selected behavioral differences be- tween modern domesticated cattle breeds: Lidia cattle (Spanish Fighting Bulls), which have been artificially selected for agonistic behaviors, display differential signals of selection on GRIK3 when compared to tame breeds [55]. These allelic differences, in turn, implicate ancient signals of selection detected on GRIK3 in the emergence of the domestic phenotype in cattle [41], resulting from selection for tameness. Similar functional implications are likely for yaks, for whom signals on GRIK3 have been detected from cattle introgression. GRIK3 has also been identified in various dog domestication studies (see Table 2). As in cattle, GRIK3 is a strong candidate for having been subject to changes in the ini- tial stages of domestication [56]. In humans, GRIK3 deletion has been implicated in severe developmental delay affecting language, motor, social, and attentional skills [57], as well as a a range of neuropsychiatric disorders and related phenotypical traits. These in- clude schizophrenia [58], bipolar disorder (manic-depressive illness) [59], major depression [60] (including significantly higher expression levels in the dorso- lateral prefrontal cortex in cases of suicide [61]), high anxiety comorbid with depression [62], increased startle response [63], and personality traits such as decreased cooperativeness and compassion, and increased shyness and harm avoidance [64]. GRIK2 has been implicated in many of the same neuropsychiatric and neu- rodevelopmental disorders as GRIK3, including schizophrenia [65, 66], bipolar disorder [67], autism spectrum disorder [68], and major depression (including emergent suicidal ideation during treatment for the disorder) [69, 61]. Both genes code for low-affinity kainate receptor subunits which share many brain- region and synaptic localizations, and which can coassemble with each other or with high-affinity kainate receptor subunits [70, 71, 72]. These similarities likely explain why GRIK2 and GRIK3 are implicated in many of the same clinical disorders, and suggests that changes to these genes may have played similar

4 roles in domestication and recent human evolution. Deletions and loss-of-function mutations on GRIK2 can cause intellectual disability and social impairments, including autistic behaviors [73, 74, 75]. A gain-of-function mutation on GRIK2 in humans has been shown to bring about syndromic changes during development that include slight microcephaly, intel- lectual disability, happy demeanor, short attention span, increased drooling, stereotyped behavior, and increased aversion to loud noises [76]. Some of these symptoms are comparable to behaviors and phenotypes often observable in do- mestic dogs. The Czechoslovakian Wolfdog, a tame hybrid resulting from a recent wolf- dog crossing experiment, has accumulated a significantly higher number of dog- like polymorphisms on GRIK2 [77], making this gene a strong candidate gene in enabling the retention of tame behaviors. Domestic rabbits — which have signals of selection on GRIK2 — exhibit gross brain changes in regions known to mediate fear response [78]. GRIK2 knockout mice experience behaviors reminiscent of manic episodes in humans, including decreased manifestations of fear, anxiety and despair in re- sponse to stressful situations, as well as increased risk-taking and aggressiveness towards conspecifics [79, 67]. Among humans, aggression and antisocial person- ality disorder are significantly higher in bipolar and schizophrenic patients than in the wider population [80, 81, 82, 83]. Polymorphisms on GRIK2 have been associated with obsessive compulsive disorder (OCD), a stress disorder, but there is no strong evidence for the in- volvement of GRIK3 as yet [84, 85]. Mice that have all five kainate receptors knocked out exhibit increased preservative and diminished exploratory behav- iors, a tendency typical in OCD [22]. Significantly lower expression of GRIK5 has been reported in the hippocam- pus, prefrontal cortex, and various thalamic nuclei of schizophrenia patients, [65, 86, 87]. A single nucleotide polymorphism (SNP) on this gene has been implicated in bipolar and panic disorders, and rare missense mutations have been associated with autism spectrum disorder [88, 89]

Metabotropic receptor genes in human and domesticated phenotypes There is ample evidence that metabotropic glutamate receptors are implicated in the emergence of tame and prosocial behaviors, in modulating the stress response, and in many of the same neurodevelopmental and neuropsychiatric disorders as kainate receptors. A fixed missense mutation on GRM3 differentiates aggressive farm-bred foxes from their tame counterparts and is thought to be one of the most impor- tant signals of the behavioral differences between the two groups [90]. There is experimental evidence that GRM3 activation in the medial prefrontal cortex is necessary for fear extinction via long-term depression of synaptic connections [91], and agonism of Group II metabotropic receptors inhibits the expression of fear potentiated startle response in rats [92]. Group II metabotropic receptors

5 are implicated in stress and mood disorders including major depression, anxi- ety disorders, and obsessive compulsive disorder, as well as schizophrenia and bipolar disorder [93, 94, 95, 96, 97, 98]. GRM3 has been identified as having longer homozygous segments in autism spectrum disorder (ASD) subjects than their parents [99], and activation of Group II receptors can help to ameliorate ASD-like social deficits in mice [100]. Agonism of Group II receptors also serves to attenuate attention deficits and impulsivity brought about by overactivation of the 5HT2A serotonin receptor in a rodent model of ADHD [101]. The Group III metabotropic receptor gene GRM8 is detected in more domes- ticate or human selective sweep studies than any other metabotropic receptor gene. In one natural experiment, very much relevant to domestication but not encompassed by these selective sweep studies, the endangered and highly inbred Apennine brown bear, known for its docility towards humans, has significantly lower nucleotide variability on GRM7 and GRM8 than the genome-wide av- erage. Similar patterns were detected on other genes related to tameness in domesticated species. The authors of the study postulated that these changes were the result of selection on reduced aggressiveness in the Apennine bear population [102]. In humans, polymorphisms on Group III receptors are often associated with mood and stress disorders, including anxiety and major depression [93, 103, 104, 105]. Studies in rodents broadly seem to support the view that activation of GRM8 serves primarily to reduce anxiety, whereas GRM7 activation has the opposite effect [93]. GRM8 knockout mice have generally increased levels of anxiety, although some measures are age- and sex-dependent, and social anxiety is attenuated by knockout [106, 107]. On the other hand, GRM7 knockout broadly decreases anxiety in mice [108]. Knockout of GRM7 causes fear-response and fear-memory deficits, while agonism of GRM8 receptors in the amygdala inhibits acquisition and expression of fear as measured by startle response [109, 110]. Polymorphisms or partial duplications on GRM7 and GRM8 have been associated with schizophrenia, ADHD, major depressive disorder, and autism spectrum disorder [111, 112, 113, 114, 115, 103, 116, 117]. GRM7 alone has been associated with bipolar disorder, while GRM8 has been implicated in severe intellectual disability [118, 119]. Polymorphisms on GRM4 have been associated with bipolar disorder, schizo- phrenia, and major depressive disorder [120, 105]. Activation of this receptor subunit can relieve autism-like symptoms and reduce anxiety-like behaviors in mice, while knockout enhances fear-conditioning responses and increases anxiety in adult mice [121, 122]. Potentiation of GRM4 receptors reduces certain mea- sures of impulsivity, while potentiating others and impairing visual attention (ADHD-like phenotype) in rats [123]. ADHD is often comorbid with obsessive compulsive behaviors and Tourette’s syndrome, both of which have been linked to aberrant glutamatergic signaling, the former being associated with both kainate and Group II metabotropic re- ceptors [124, 125]. Group III metabotropic receptors have been proposed as potential targets for treatment of OCD and Tourette’s syndrome [126].

6 S3 Glutamate receptors in the CNS Kainate receptor expression Although kainate receptors are structurally similar to AMPA receptors, “they fulfill a more varied range of synaptic and extrasynaptic roles” than their iono- tropic counterparts [26]. Apart from ligand gating, kainate receptors modulate glutamate release and activate second-messenger signaling cascades to a greater extent than AMPA or NMDA receptors [127, 128, 129]. Kainate receptors may serve to facilitate or inhibit glutamate or GABA release, regulate dendritic outgrowth during development, potentiate AMPA-dependent excitatory synap- tic transmission, or activate distinct “non-canonical” (non-ionotropic) signal- ing, including Gi/o proteins, the principal second-messenger signaling targets of Group II and III metabotropic receptors [130, 131, 132, 129]. Thus, depending on the primary roles played by kainate receptor subunits in a given brain region, their expression may have distinct functional implications far beyond excitatory synaptic transmission [129]. Here, we focus on the regional expression of the low affinity receptor genes, GRIK2 (because of signals of potential convergence among domesticates) and GRIK3 (because of parallel signals among domesticates and humans), as well as the high-affinity receptor gene GRIK5 (given signals of selection detected in AMH). Because signals of convergence with domesticates have yet to be identi- fied for GRIK5, this receptor gene received little attention in the cross-species comparisons of selective sweep studies. However, GRIK5 has been identified in both selective-sweep studies of modern human populations and among high- frequency differences distinguishing archaic from modern human populations [6, 50]. For these reasons we include it in our review of expression data. GRIK2 mRNA is highly expressed in cerebellar granule cells and is also found in the caudate putamen and hippocampus, while GRIK3 is strongest expressed in deep layers of the cerebral cortex, the cingulate cortex, the subicu- lum, the caudate-putamen, thalamic reticular nucleus, and stellate/basket cells of the cerebellum [133, 72]. GRIK5 is the most universally expressed of all kainate receptor genes and can be detected across most brain regions, although the encoded subunit must coassemble with low-affinity kainate receptor sub- units (GRIK1-GRIK3) in order to form functional ligand-gated ion channels [134, 133, 72]. In general, kainate receptor is highest in the hip- pocampus and the cerebellum [72]. Kainate receptor mRNA is detectable in the amygdala, although GRIK1 is considerably more abundant than other receptor genes that have been detected, with GRIK2 having moderate and GRIK5 low expression [135]. Expression levels of kainate receptor genes are developmentally regulated, peaking in the rat brain in the late embryonic and early postnatal period [133]. As early as embryonic day 10, four kainate receptor genes (GRIK2-GRIK5 ) are expressed in the rat neural tube. This expression is higher than that of AMPA receptors and is present in both progenitor cells and differentiated neurons, with GRIK2 and GRIK5 subunits being upregulated during neural cell differentia-

7 tion [136]. During later embryonic periods, brain expression is highest in the cerebellum and striatum, later extending to the cortex, septum, hippocampus, and thalamus. Expression levels drop markedly in the thalamus from the late postnatal period onwards [133]. GRIK5 is highest expressed in the cortical plate in the late embryonic period. In general, GRIK2 is not highly expressed in the cortex, although it is abundant in the cingulate cortex, peaking during early postnatal development. GRIK3 is more broadly expressed in the surrounding cortex than GRIK2, but autoradiograph images nonetheless show increased ex- pression in the cingulate cortex during the immediate postnatal period, and moderate to abundant expression into adulthood [134, 133]. Highest expression levels for GRIK3 during postnatal development are detected in the entorhinal cortex [133]. In the rat hippocampus, expression of kainate receptor genes peaks in the late embryonic and early postnatal period. This is particularly transient in CA1 but remains high in CA3 into adulthood. Expression of kainate receptor genes in the dentate gyrus (DG) also remains high from the postnatal period into adulthood. Similarly, immunolabeling of GRIK2, GRIK3, and GRIK5 sub- units in adult rats show highest abundance in the pyramidal layer of CA3 and in neurons in DG, being present in both cell bodies and apical dendrites [70]. GRIK3 and GRIK5 are moderately expressed in the septum from embryonic development, with these levels remaining unchanged in the lateral septum into adulthood [133, 134]. GRIK2, GRIK3, and GRIK5 are the only kainate receptor genes consistently detected in the striatum during embryonic development, with GRIK5 remaining abundant into adulthood [133]. These genes are moderate to abundantly expressed in the thalamus at different stages of prenatal devel- opment, while GRIK3 expression remains strong in the reticular nucleus into adulthood. Finally, in the cerebellum, GRIK2 and GRIK5 are the predominant kainate receptor genes expressed in granule cells throughout life, while GRIK3 is the only kainate receptor gene expressed in stellate/basket cells [134, 133]. In a study of the human fetal cortex, GRIK3 and GRIK5 mRNA were far more abundant than those of other kainate receptors throughout the different developmental stages measured (between gestation week 8 and week 20) [137]. This broadly matches with data from the prenatal rat brain, in which GRIK2, GRIK3, and GRIK5 are the most widely expressed subunits [133]. Moreover, both the expression of receptor mRNA and receptor binding — suggesting full translation into functional subunits — was found to be greater for kainate recep- tors during fetal development than for any other ionotropic glutamate receptors, especially during the later gestational periods measured [137]. The expression of kainate receptor transcripts is highly synchronized with that of NMDA re- ceptors at each of the different stages measured. It is likely that these subunits are involved in modulating different processes of neurogenesis during ontogeny, including neuronal migration [137]. Somewhat contrary these findings, AMPA receptor binding from gestational week 16.5 to week 26 has been found to be considerably denser in the hip- pocampus, entorhinal cortex, temporal cortex, cingulate cortex, putamen, and thalamus than for than for the other ionotropic receptors [138]. NMDA recep-

8 tor densities were also greater than those of kainate receptors, although these were broadly comparable across most regions and time points, perhaps due to the same synchrony observed in other studies [138, 137]. These discrepancies between mRNA measures and receptor binding densities may be down to delays of the translation into full kainate and NMDA receptor subunits in the regions studied [137]. Some evidence for this delayed development of receptor subunits comes from comparisons of receptor binding in the hippocampi of second trimester fetuses with postmortem measures of full-term infants, three-month-olds, and adults: NMDA receptors made up around half of total glutamate receptors in the in- fant hippocampi measured, as compared to around a third during the second trimester of gestation or in adults. This was especially the case in the stratum lucidum (CA3) and the molecular layer of the fascia dentata (DG). Moreover, the percentage of the remaining glutamate receptors that bound quisqualate was minimal throughout development [139]. Given that quisqualate binds AMPA receptors with much higher affinity than kainate receptors, a considerable per- centage of the remaining glutamate receptors may be expected to be kainate preferring [140, 141, 139]. A separate study of kainate binding in the newborn hippocampus confirms this, with a marked increase relative to prenatal or adult levels in the stratum lucidum and the molecular layer of the fascia dentata, again suggesting synchrony between kainate and NMDA receptor expression during development [142]. Given the relative abundance of GRIK3 and GRIK5 ex- pression during earlier stages of human fetal development, it is not implausible that these receptor genes also drive the increase in postnatal expression. Postmortem mRNA measures taken from adult human brains confirm GRIK2, GRIK3, and GRIK5 as the highest expressed kainate receptor genes. However, lower relative expression patterns have been detected for GRIK3 in adult hu- mans compared to adult rat or fetal human brains. Overall, the dentate gyrus and CA3 remain regions of highest expression, with GRIK2 and GRIK5 pre- dominant [65]. It should be noted that studies of adult mRNA expression and receptor binding generally find decreased densities in the human hippocampus relative to rodents and monkeys [143, 144]. However, contrary to the findings of the human study mentioned above [65], it has been posited that these decreases are primarily due to lower expression of GRIK2 and GRIK4, rather than GRIK3 and GRIK5 [143]. Moreover, during the postnatal peak of kainate binding in CA3 and DG, human densities of kainate receptors are comparable to or even exceed those of the rat [142]. Possibly, then, selective pressures in our lineage may have driven the increased ontogenetic expression of GRIK3 and GRIK5. Non-selective immunolabeling of GRIK1-GRIK3 in the monkey hippocam- pus suggest more qualitative differences in localization as compared with both human and rat data. These low-affinity subunits were found to be densest in CA1 and the subiculum rather than DG or CA3 [145]. The authors of this study suggest that discrepancies identified between the monkey and the rat may be down to species differences. By comparison, a similar postmortem study in humans found strongest labeling of these subunits in CA2 and CA3 pyrami- dal neurons (CA2 most abundantly) [146]. Again, it cannot be ruled out that

9 human-monkey differences in low-affinity kainate receptor expression are driven in part by the regulatory changes on GRIK3 that came under selection in our lineage. In histological studies of the rat hypothalamus, GRIK2, GRIK3, and GRIK5 receptor proteins have been detected at moderate levels in the supraoptic nu- cleus (SON), arcuate nucleus, and the junction of the median eminence and infundibulum. Light to moderate staining was detected in the retrochiasmatic region of the SON and the medial mammillary nucleus [70]. Another study found that non-NMDA (mainly kainate) receptors were bound most predom- inantly in the SON, anterior hypothalamic area, and paraventricular nucleus (PVN) [147]. Both GRIK2 and GRIK5 subunit mRNAs are expressed in the anterior parvocellular region of the PVN, while GRIK5 is highest expressed in the medial parvocellular nucleus and is expressed on CRH-releasing neurons [148]. GRIK2 and GRIK3 are most highly expressed in the the subparaventric- ular zone and perinuclear region surrounding the PVN [149]. Studies in vitro confirm that kainate receptors are extensively expressed in the hypothalamus during embryonic development [150], although we are unaware of any study detailing specific subunit abundances or localizations during ontogeny. These expression data underscore the particular importance of kainate recep- tor genes that most often show signals of selection in domesticates and modern humans. Evidence that GRIK3 and GRIK5 are the kainate receptor subunits with highest expression levels in the developing human brain is suggestive that they are important for neurogenesis and differentiation. Although no easy com- parisons can be made between the brain-expression data for embryonic or peri- natal rats and a human fetuses, GRIK3 and GRIK5 expression levels are high in each species, suggesting that similar developmental processes are regulated by these subunits. The comparatively higher expression of GRIK2 as compared with GRIK3 in the rat suggests there may be a more prominent role for this gene in CNS development in mammals other than humans. If this is so, hu- man GRIK3 may occupy homologous ontogenetic functions to GRIK2, perhaps helping to explain the tendency for both GRIK2 and GRIK3 to show signals of selection in domesticated species, whereas this is only the case for GRIK3 in AMH. The relative abundance of kainate receptor gene expression in limbic struc- tures (e.g. hippocampus, entorhinal cortex, cingulate cortex, and, to a lesser extent, the lateral septum and hypothalamus) points to the importance of these genes in the primary functions of these structures, including control of emo- tions, memory, learning, and neuroendocrine modulation. This is especially the case during the postnatal period, when kainate receptor genes are highest ex- pressed. The cingulate cortex and hippocampus, which prominently modulate HPA-feedback activity, retain moderate to high expression of these genes into adulthood. As detailed above, in the DG and CA3, GRIK2 and GRIK5 are expressed most abundantly in the adult rat brain, while GRIK3 and GRIK5 are abundant in DG during early postnatal development.

10 Metabotropic expression Group II and Group III metabotropic glutamate receptor genes are expressed throughout the central nervous system and are often highly expressed in limbic structures, including the hippocampal formation, as well as in sensory pathways, such as the olfactory and visual systems [72]. The Group II receptor genes, GRM2 and GRM3 are moderate to abundantly expressed in various regions of the limbic cortex, Golgi cells of the cerebellum, the dentate gyrus, and in distinct amygdaloid nuclei [151, 152]. Of the Group III receptor genes, GRM4, GRM7, and GRM8 are distributed widely across the brain, GRM7 most expansively and abundantly, followed by GRM4, and GRM8, which is widely distributed, but expressed in lower quantities [153, 154]. GRM6 expression is limited to the ON-bipolar cells of the retina. Group II and III receptors play a more prominent role than Group I in the presynaptic modulation of glutamatergic signaling [72, 154]. In the rat brain, GRM2 is most intensely expressed in the in Golgi cells of the cerebellum, accessory olfactory bulb, and anterior olfactory nucleus, followed by, the entorhinal and parasubicular cortices, granule cells of the dentate gyrus, basolateral and basomedial amygdala, various thalamic nuclei, the medial mam- millary nucleus of the hypothalamus, and the cingulate and retrosplenial cortices [151]. Human GRM2 mRNA is most prominently detected in the cerebellum, hypothalamus, and thalamus, with lighter expression in the hippocampus [155]. In the rat, GRM3 expression is most abundant in the thalamic reticular nucleus, followed by the lateral and basolateral amygdala, Golgi cells of the cerebellum, the cingulate, retrosplenial, perirhinal, entorhinal, and parasubic- ular cortices, distinct subparts of the brain stem, and the supraoptic nucleus of the hypothalamus, with lighter expression in the paraventricular and lateral areas of the hypothalamus, as well as granule cells of the dentate gyrus, and stellate/basket cells of the cerebellum [152]. In humans, GRM3 mRNA has been shown to be similarly expressed in the dentate gyrus, cerebellum, and thalamic reticular nucleus, although there is also evidence of more extensive expression across thalamic nuclei and cerebellar cell types. Strong GRM3 receptor im- munoreactivity signals have been detected in the human prefrontal cortex, and low to moderate levels in the hippocampus, amygdala, and thalamus [156]. Non-selective labeling of Group II receptors in the human hippocampus iden- tified dense staining in the dentate gyrus, CA2-CA4, and granule cells of CA3, with lighter staining in CA1 and the subicular cortex [157]. Elsewhere in the brain Group II immunoreactivity is present in the prefrontal cortex and striatum [156] GRM4 is highest expressed in the rat sensory ganglia and in granule cells of the cerebellum. In general, it is more extensively expressed than both Group II receptors in the olfactory system and septum, but less so in the limbic cortices. Expression in the amygdala is limited to the rostral portion of the interca- lated nuclei. Like GRM2, expression of GRM4 within the rat hypothalamus is mostly limited to the medial mammillary nucleus. Moderate expression within the hippocampus is mostly limited to the rat dentate gyrus, with similar levels

11 detected in the entorhinal cortex [153]. However, immunocytochemical local- ization has detected more intense labeling of GRM4 proteins on cell bodies and apical dendrites in CA1 and CA3, and on the bodies of granule cells in DG [158]. In humans, GRM4 mRNA is strongest detected in the cerebellum, and more moderately in the hippocampus, hypothalamus, and thalamus [159]. In a postmortem human immunohistochemical study of the hippocampus, a specific GRM4 isoform showed moderate labeling on CA3 mossy fibres, with weaker la- beling in CA2. Labeling was also detected on the hippocampal efferents of the alveus, parts of which contribute to inputs of the lateral septal nuclei [157, 160]. These data largely parallel with data from the rat, although there is evidence of higher GRM4 mRNA levels in the human caudate nucleus and putamen, which form part of the dorsal striatum [161]. GRM7 is expressed in almost all brain regions in the rat, generally at mod- erate levels. This includes all limbic, amygdalar, hippocampal, and thalamic regions, and practically all hypothalamic subdivisions. Like GRM4, intense im- munocytochemical staining can be detected for GRM7 subunits in all regions of the hippocampus, although this is most apparent in neuropil rather than on cell bodies [158]. Highest gene expression is detected in the sensory ganglia, the coeruleus within the brainstem, the main olfactory bulb, and the me- dial septal nucleus [153]. In accordance with studies of the rat, human GRM7 mRNA levels are also widely distributed at moderate levels in various regions of the human brain, being highest in the neocortex, cerebellum, and regions DG and CA3 of the hippocampus, followed by CA1 [162, 163]. Expression was also detected in the thalamus, with lower levels in the caudate-putamen, in contrast to studies of the rat and the inverse of species expression differences for GRM4 [163]. In the rat GRM8 is highest expressed in the olfactory bulb, pontine nuclei of the brainstem, piriform cortex, and reticular nucleus of the thalamus, with lower expression in the the neocortex, hippocampus, basolateral amygdala, and mammillary body of the hypothalamus [164, 165, 162]. In humans, distinct GRM8 isoforms are differentially but widely expressed, with the most abun- dant variant showing highest expression in the thalamus, subthalamic nucleus, caudate nucleus, and amygdala, with a second variant being expressed in the visual cortex, caudate-putamen, and cerebellum [166]. Among all glutamate receptor subtypes, metabotropic glutamate receptor binding densities in the rat hypothalamus are second only to NMDA receptors (particularly in dorsomedial, ventromedial, and paraventricular regions) [147]. Given the mRNA expression levels reviewed above and relatively low Group I metabotropic receptor expression in the adult rat hypothalamus, a considerable percentage of these metabotropic receptors may be expected to comprise mul- tiple Group II and III subtypes, most prominently in the medial mammillary nucleus [167, 151, 152, 153]. GRM2 and GRM3 receptor proteins have been detected in the mouse and rat hypothalami and have been shown to modulate HPA function [168]. Unlike kainate receptors, developmental regulation of Group II and III meta- botropic receptors in the rat hypothalamus may tend towards increased ex-

12 pression in adulthood. Such increases have been identified in both arcuate and suprachiasmatic nuclei for Group II and III subunits [169]. On the other hand, across brain regions, most metabotropic receptors show similar patterns to kainate receptors, peaking in the early postnatal period. GRM2 shows a progressive increase in expression during postnatal development in the rat, but remains high in the entorhinal cortex, DG, and various nuclei of the thalamus into adulthood. Low expression levels were found in CA1 and CA3 (in con- trast to the human postmortem study described above [157]), and none was detected in the striatum [170]. In the human fetal brain GRM2 expression is higher than in that of adults, although no regional-specific prenatal expression has been described [155]. GRM3 generally has highest postnatal expression at birth, especially in the striatum and cerebellum, decreasing progressively throughout development, but remaining high in the thalamic reticular nucleus into adulthood [170, 171]. Expression levels for GRM4 were found to be low throughout development in one study of the rat, with little change into adulthood, except in granule cells of the cerebellum, where they rise considerably [170]. Other studies have found a similar rise in GRM4 expression on cell bodies in all major regions of the hippocampus in adulthood [171]. GRM7 proteins show increased postnatal abundance in the rat relative to adult levels across almost all regions studied, including the pons, cerebellum, thalamus, hippocampus, olfactory cortex, and striatum, although expression remains relatively high in the hippocampus, bed nucleus of stria terminalis (BNST), medial mammillary body of the hypotha- lamus, and various thalamic nuclei into adulthood [172]. In humans, GRM8 shows increased expression in the fetal brain than in that of adults [166]. Kainate and Group II and III metabotropic receptors have broadly overlap- ping localizations and timings of expression, with predominant postnatal expres- sion in limbic cortices, the hippocampus (especially DG and CA3), cerebellum, thalamus (particularly the reticular nucleus), and hypothalamus (most notably the medial mammillary nucleus, although perhaps asynchronously). Differences include higher metabotropic expression in sensory pathways, as well as the amygdala, thalamus, distinct hippocampal regions, and in the adult hypothala- mus more generally. Kainate receptor expression seems to be more predominant in the cingulate cortex and striatum, particularly during ontogeny. Both recep- tor subtypes have metabotropic functions modulating the release of glutamate, and both are part of G-protein coupled signaling cascades. Together, these re- ceptor subtypes are extensively expressed in regions crucial for the modualtion of how an organism deals with stress, from the moment sensory inputs for a stressor are received right through to regulation of feedback mechanisms that inform best responses.

13 References

[1] Fernando Racimo. Testing for Ancient Selection Using Cross-population Allele Frequency Differentiation. Genetics, 202(2):733–750, February 2016. [2] Ivica Medugorac, Alexander Graf, C´ecileGrohs, Sophie Rothammer, Yon- don Zagdsuren, Elena Gladyr, Natalia Zinovieva, Johanna Barbieri, Doris Seichter, Ingolf Russ, Andr´eEggen, Garrett Hellenthal, Gottfried Brem, Helmut Blum, Stefan Krebs, and Aur´elienCapitan. Whole-genome analy- sis of introgressive hybridization and characterization of the bovine legacy of Mongolian yaks. Nature Genetics, 49(3):470–475, March 2017. [3] Christoph Straub, Wei Zhang, and James R. Howe. Neto2 Modulation of Kainate Receptors with Different Subunit Compositions. Journal of Neuroscience, 31(22):8078–8082, June 2011. [4] Man Tang, Evgueni Ivakine, Vivek Mahadevan, Michael W. Salter, and Roderick R. McInnes. Neto2 Interacts with the Scaffolding Protein GRIP and Regulates Synaptic Abundance of Kainate Receptors. PLOS ONE, 7(12):e51433, June 2012. [5] Jessica C. Pressey, Vivek Mahadevan, C. Sahara Khademullah, Zahra Dar- gaei, Jonah Chevrier, Wenqing Ye, Michelle Huang, Alamjeet K. Chauhan, Steven J. Meas, Pavel Uvarov, Matti S. Airaksinen, and Melanie A. Woodin. A kainate receptor subunit promotes the recycling of the - specific K+-Cl- co-transporter KCC2 in hippocampal neurons. Journal of Biological Chemistry, 292(15):6190–6201, April 2017. [6] St´ephanePeyr´egne,Michael James Boyle, Michael Dannemann, and Kay Pr¨ufer. Detecting ancient positive selection in humans using extended lineage sorting. Genome Research, 27(9):1563–1572, September 2017. [7] Gr´egoryGauvain, Ingrid Chamma, Quentin Chevy, Carolina Cabezas, Theano Irinopoulou, Natalia Bodrug, Mich`ele Carnaud, Sabine L´evi, and Jean Christophe Poncer. The neuronal K-Cl cotransporter KCC2 influences postsynaptic AMPA receptor content and lateral diffusion in dendritic spines. Proceedings of the National Academy of Sciences, 108(37):15474–15479, September 2011. [8] Evgueni A. Ivakine, Brooke A. Acton, Vivek Mahadevan, Jake Ormond, Man Tang, Jessica C. Pressey, Michelle Y. Huang, David Ng, Eric Delpire, Michael W. Salter, Melanie A. Woodin, and Roderick R. McInnes. Neto2 is a KCC2 interacting protein required for neuronal Cl- regulation in hip- pocampal neurons. Proceedings of the National Academy of Sciences, page 201212907, February 2013. [9] Ran Tao, Chao Li, Erin N. Newburn, Tianzhang Ye, Barbara K. Lip- ska, Mary M. Herman, Daniel R. Weinberger, Joel E. Kleinman, and

14 Thomas M. Hyde. Transcript-Specific Associations of SLC12a5 (KCC2) in Human Prefrontal Cortex with Development, Schizophrenia, and Af- fective Disorders. Journal of Neuroscience, 32(15):5216–5222, April 2012. [10] Nancy D. Merner, Madison R. Chandler, Cynthia Bourassa, Bo Liang, Arjun R. Khanna, Patrick Dion, Guy A. Rouleau, and Kristopher T. Kahle. Regulatory domain or CpG site variation in SLC12a5, encoding the chloride transporter KCC2, in human autism and schizophrenia. Frontiers in Cellular Neuroscience, 9, 2015. [11] Tommy St¨odberg, Amy McTague, Arnaud J. Ruiz, Hiromi Hirata, Juan Zhen, Philip Long, Irene Farabella, Esther Meyer, Atsuo Kawahara, Grace Vassallo, Stavros M. Stivaros, Magnus K. Bjursell, Henrik Stran- neheim, Stephanie Tigerschi¨old,Bengt Persson, Iftikhar Bangash, Kr- ishna Das, Deborah Hughes, Nicole Lesko, Joakim Lundeberg, Rod C. Scott, Annapurna Poduri, Ingrid E. Scheffer, Holly Smith, Paul Gissen, Stephanie Schorge, Maarten E. A. Reith, Maya Topf, Dimitri M. Kull- mann, Robert J. Harvey, Anna Wedell, and Manju A. Kurian. Mutations in SLC12A5 in epilepsy of infancy with migrating focal seizures. Nature Communications, 6:8038, September 2015. [12] Hirotomo Saitsu, Miho Watanabe, Tenpei Akita, Chihiro Ohba, Kenji Sugai, Winnie Peitee Ong, Hideaki Shiraishi, Shota Yuasa, Hiroshi Mat- sumoto, Khoo Teik Beng, Shinji Saitoh, Satoko Miyatake, Mitsuko Nakashima, Noriko Miyake, Mitsuhiro Kato, Atsuo Fukuda, and Naomichi Matsumoto. Impaired neuronal KCC2 function by biallelic SLC12A5 mu- tations in migrating focal seizures and severe developmental delay. Scien- tific Reports, 6:30072, July 2016. [13] Henrike O. Heyne, Susann Lautenschl¨ager, Ronald Nelson, Fran¸cois Besnier, Maxime Rotival, Alexander Cagan, Rimma Kozhemyakina, Irina Z. Plyusnina, Lyudmila Trut, Orjan¨ Carlborg, Enrico Petretto, Leonid Kruglyak, Svante P¨a¨abo, Torsten Sch¨oneberg, and Frank W. Al- bert. Genetic Influences on Brain Gene Expression in Rats Selected for Tameness and Aggression. Genetics, 198(3):1277–1290, November 2014. [14] Swapan Mallick, Heng Li, Mark Lipson, Iain Mathieson, Melissa Gym- rek, Fernando Racimo, Mengyao Zhao, Niru Chennagiri, Susanne Nor- denfelt, Arti Tandon, Pontus Skoglund, Iosif Lazaridis, Sriram Sankarara- man, Qiaomei Fu, Nadin Rohland, Gabriel Renaud, Yaniv Erlich, Thomas Willems, Carla Gallo, Jeffrey P. Spence, Yun S. Song, Giovanni Po- letti, Francois Balloux, George van Driem, Peter de Knijff, Irene Gal- lego Romero, Aashish R. Jha, Doron M. Behar, Claudio M. Bravi, Cristian Capelli, Tor Hervig, Andres Moreno-Estrada, Olga L. Posukh, Elena Balanovska, Oleg Balanovsky, Sena Karachanak-Yankova, Hov- hannes Sahakyan, Draga Toncheva, Levon Yepiskoposyan, Chris Tyler- Smith, Yali Xue, M. Syafiq Abdullah, Andres Ruiz-Linares, Cynthia M. Beall, Anna Di Rienzo, Choongwon Jeong, Elena B. Starikovskaya, Ene

15 Metspalu, J¨uriParik, Richard Villems, Brenna M. Henn, Ugur Hodoglugil, Robert Mahley, Antti Sajantila, George Stamatoyannopoulos, Joseph T. S. Wee, Rita Khusainova, Elza Khusnutdinova, Sergey Litvinov, George Ayodo, David Comas, Michael F. Hammer, Toomas Kivisild, William Klitz, Cheryl A. Winkler, Damian Labuda, Michael Bamshad, Lynn B. Jorde, Sarah A. Tishkoff, W. Scott Watkins, Mait Metspalu, Stanislav Dryomov, Rem Sukernik, Lalji Singh, Kumarasamy Thangaraj, Svante P¨a¨abo, Janet Kelso, Nick Patterson, and David Reich. The Simons Genome Diversity Project: 300 genomes from 142 diverse populations. Nature, 538(7624):201–206, October 2016. [15] S. E. Stewart, D. Yu, J. M. Scharf, B. M. Neale, J. A. Fagerness, C. A. Mathews, P. D. Arnold, P. D. Evans, E. R. Gamazon, L. Osiecki, L. McGrath, S. Haddad, J. Crane, D. Hezel, C. Illman, C. Mayerfeld, A. Konkashbaev, C. Liu, A. Pluzhnikov, A. Tikhomirov, C. K. Edlund, S. L. Rauch, R. Moessner, P. Falkai, W. Maier, S. Ruhrmann, H.-J. Grabe, L. Lennertz, M. Wagner, L. Bellodi, M. C. Cavallini, M. A. Richter, E. H. Cook Jr, J. L. Kennedy, D. Rosenberg, D. J. Stein, S. M. J. Hemmings, C. Lochner, A. Azzam, D. A. Chavira, E. Fournier, H. Garrido, B. Sheppard, P. Uma˜na,D. L. Murphy, J. R. Wendland, J. Veenstra-VanderWeele, D. Denys, R. Blom, D. Deforce, F. Van Nieuwer- burgh, H. G. M. Westenberg, S. Walitza, K. Egberts, T. Renner, E. C. Miguel, C. Cappi, A. G. Hounie, M. Concei¸c˜aodo Ros´ario,A. S. Sam- paio, H. Vallada, H. Nicolini, N. Lanzagorta, B. Camarena, R. Delorme, M. Leboyer, C. N. Pato, M. T. Pato, E. Voyiaziakis, P. Heutink, D. C. Cath, D. Posthuma, J. H. Smit, J. Samuels, O. J. Bienvenu, B. Cullen, A. J. Fyer, M. A. Grados, B. D. Greenberg, J. T. McCracken, M. A. Rid- dle, Y. Wang, V. Coric, J. F. Leckman, M. Bloch, C. Pittenger, V. Eapen, D. W. Black, R. A. Ophoff, E. Strengman, D. Cusi, M. Turiel, F. Frau, F. Macciardi, J. R. Gibbs, M. R. Cookson, A. Singleton, North American Brain Expression Consortium, S. Arepalli, M. R. Cookson, A. Dillman, L. Ferrucci, J. R. Gibbs, D. G. Hernandez, R. Johnson, D. L. Longo, M. A. Nalls, R. O’Brien, A. Singleton, B. Traynor, J. Troncoso, M. van der Brug, H. R. Zielke, A. Zonderman, J. Hardy, UK Brain Expression Database, J. A. Hardy, M. Ryten, C. Smith, D. Trabzuni, R. Walker, Mike Weale, A. T. Crenshaw, M. A. Parkin, D. B. Mirel, D. V. Conti, S. Purcell, G. Nestadt, G. L. Hanna, M. A. Jenike, J. A. Knowles, N. Cox, and D. L. Pauls. Genome-wide association study of obsessive-compulsive disorder. Molecular Psychiatry, 18(7):788–798, July 2013. [16] Jiang Li, Jiajia Cui, Xiuhai Wang, Jianhua Ma, Haitao Niu, Xu Ma, Xinhua Zhang, and Shiguo Liu. An association study between DL- GAP1 rs11081062 and EFNA5 rs26728 polymorphisms with obses- sive–compulsive disorder in a Chinese Han population. Neuropsychiatric Disease and Treatment, 11:897–905, April 2015. [17] M. Mattheisen, J. F. Samuels, Y. Wang, B. D. Greenberg, A. J. Fyer,

16 J. T. McCracken, D. A. Geller, D. L. Murphy, J. A. Knowles, M. A. Gra- dos, M. A. Riddle, S. A. Rasmussen, N. C. McLaughlin, E. L. Nurmi, K. D. Askland, H.-D. Qin, B. A. Cullen, J. Piacentini, D. L. Pauls, O. J. Bienvenu, S. E. Stewart, K.-Y. Liang, F. S. Goes, B. Maher, A. E. Pulver, Y. Y. Shugart, D. Valle, C. Lange, and G. Nestadt. Genome-wide asso- ciation study in obsessive-compulsive disorder: results from the OCGAS. Molecular Psychiatry, 20(3):337–344, March 2015. [18] Mikkel Schubert, H´akon J´onsson,Dan Chang, Clio Der Sarkissian, Luca Ermini, Aur´elien Ginolhac, Anders Albrechtsen, Isabelle Dupanloup, Adrien Foucal, Bent Petersen, Matteo Fumagalli, Maanasa Raghavan, Andaine Seguin-Orlando, Thorfinn S. Korneliussen, Amhed M. V. Ve- lazquez, Jesper Stenderup, Cindi A. Hoover, Carl-Johan Rubin, Ahmed H. Alfarhan, Saleh A. Alquraishi, Khaled A. S. Al-Rasheid, David E. MacHugh, Ted Kalbfleisch, James N. MacLeod, Edward M. Rubin, Thomas Sicheritz-Ponten, Leif Andersson, Michael Hofreiter, Tomas Marques-Bonet, M. Thomas P. Gilbert, Rasmus Nielsen, Laurent Ex- coffier, Eske Willerslev, Beth Shapiro, and Ludovic Orlando. Prehistoric genomes reveal the genetic foundation and cost of horse domestication. Proceedings of the National Academy of Sciences, 111(52):E5661–E5669, December 2014. [19] Georgia J. Mason. Stereotypies: a critical review. Animal Behaviour, 41(6):1015–1037, June 1991. [20] C. Nicol. Understanding equine stereotypies. Equine Veterinary Journal. Supplement, (28):20–25, April 1999. [21] Nicholas H Dodman, Edward I Ginns, Louis Shuster, Alice A Moon- Fanelli, Marzena Galdzicka, Jiashun Zheng, Alison L Ruhe, and Mark W Neff. Genomic Risk for Severe Canine Compulsive Disorder, a Dog Model of Human OCD. 14(1):19, January 2016. [22] Jian Xu, John J. Marshall, Herman B. Fernandes, Toshihiro Nomura, Bryan A. Copits, Daniele Procissi, Susumu Mori, Lei Wang, Yongling Zhu, Geoffrey T. Swanson, and Anis Contractor. Complete disruption of the kainate receptor gene family results in corticostriatal dysfunction in mice. Cell reports, 18(8):1848–1857, February 2017. [23] Elizabeth P Garcia, Sunil Mehta, Leslie A. C Blair, David G Wells, Jing Shang, Teruyuki Fukushima, Justin R Fallon, Craig C Garner, and John Marshall. SAP90 Binds and Clusters Kainate Receptors Causing Incom- plete Desensitization. Neuron, 21(4):727–739, October 1998. [24] M. P. Coba, M. J. Ramaker, E. V. Ho, S. L. Thompson, N. H. Komiyama, S. G. N. Grant, J. A. Knowles, and S. C. Dulawa. Dlgap1 knockout mice exhibit alterations of the postsynaptic density and selective reductions in sociability. Scientific Reports, 8(1):2281, February 2018.

17 [25] Jeffrey M. Welch, Jing Lu, Ramona M. Rodriguiz, Nicholas C. Trotta, Joao Peca, Jin-Dong Ding, Catia Feliciano, Meng Chen, J. Paige Adams, Jianhong Luo, Serena M. Dudek, Richard J. Weinberg, Nicole Calakos, William C. Wetsel, and Guoping Feng. Cortico-striatal synaptic defects and OCD-like behaviours in Sapap3 -mutant mice. Nature, 448(7156):894– 900, August 2007. [26] John J. Marshall, Jian Xu, and Anis Contractor. Kainate receptors inhibit glutamate release via mobilization of endocannabinoids in striatal direct pathway Spiny Projection Neurons (dSPNs). Journal of Neuroscience, pages 1788–17, March 2018.

[27] Anna V. Kukekova, Jennifer L. Johnson, Xueyan Xiang, Shaohong Feng, Shiping Liu, Halie M. Rando, Anastasiya V. Kharlamova, Yury Her- beck, Natalya A. Serdyukova, Zijun Xiong, Violetta Beklemischeva, Klaus- Peter Koepfli, Rimma G. Gulevich, Anastasiya V. Vladimirova, Jessica P. Hekman, Polina L. Perelman, Aleksander S. Graphodatsky, Stephen J. O’Brien, Xu Wang, Andrew G. Clark, Gregory M. Acland, Lyudmila N. Trut, and Guojie Zhang. Red fox genome assembly identifies genomic re- gions associated with tame and aggressive behaviours. Nature Ecology & Evolution, 2(9):1479–1491, September 2018. [28] Jeffrey N. Savas, Lu´ısF. Ribeiro, Keimpe D. Wierda, Rebecca Wright, Laura A. DeNardo-Wilke, Heather C. Rice, Ingrid Chamma, Yi-Zhi Wang, Roland Zemla, Mathieu Lavall´ee-Adam, Kristel M. Vennekens, Matthew L. O’Sullivan, Joseph K. Antonios, Elizabeth A. Hall, Olivier Thoumine, Alan D. Attie, John R. Yates, Anirvan Ghosh, and Joris de Wit. The Sorting Receptor SorCS1 Regulates Trafficking of Neurexin and AMPA Receptors. Neuron, 87(4):764–780, August 2015.

[29] St´ephanePeyr´egne,Michael James Boyle, Michael Dannemann, and Kay Pruefer. Detecting ancient positive selection in humans using extended lineage sorting. bioRxiv, December 2016. [30] S. Glerup, U. Bolcho, S. Mølgaard, S. Bøggild, C. B. Vaegter, A. H. Smith, J. L. Nieto-Gonzalez, P. L. Ovesen, L. F. Pedersen, A. N. Fjor- back, M. Kjolby, H. Login, M. M. Holm, O. M. Andersen, J. R. Nyen- gaard, T. E. Willnow, K. Jensen, and A. Nykjaer. SorCS2 is required for BDNF -dependent plasticity in the hippocampus. Molecular Psychiatry, 21(12):1740–1751, December 2016. [31] Simon Boggild, Simon Molgaard, Simon Glerup, and Jens Randel Nyen- gaard. Highly segregated localization of the functionally related vps10p receptors sortilin and SorCS2 during neurodevelopment. Journal of Com- parative Neurology, 526(8):1267–1286, June 2018. [32] Tilman Breiderhoff, Gitte B. Christiansen, Lone T. Pallesen, Christian Vaegter, Anders Nykjaer, Mai Marie Holm, Simon Glerup, and Thomas E.

18 Willnow. Sortilin-Related Receptor SORCS3 Is a Postsynaptic Modulator of Synaptic Depression and Fear Extinction. PLOS ONE, 8(9):e75006, September 2013. [33] Yan Li, Guo-Dong Wang, Ming-Shan Wang, David M. Irwin, Dong-Dong Wu, and Ya-Ping Zhang. Domestication of the Dog from the Wolf Was Promoted by Enhanced Excitatory Synaptic Plasticity: A Hypothesis. Genome Biology and Evolution, 6(11):3115–3121, November 2014. [34] Takashi Maejima, Saori Oka, Yuki Hashimotodani, Takako Ohno-Shosaku, Atsu Aiba, Dianqing Wu, Keizo Waku, Takayuki Sugiura, and Masanobu Kano. Synaptically Driven Endocannabinoid Release Requires Ca2+- Assisted Metabotropic Glutamate Receptor Subtype 1 to Phospholipase C \beta4 Signaling Cascade in the Cerebellum. Journal of Neuroscience, 25(29):6826–6835, July 2005. [35] Mark J. Rieder, Glenn E. Green, Sarah S. Park, Brendan D. Stam- per, Christopher T. Gordon, Jason M. Johnson, Christopher M. Cun- niff, Joshua D. Smith, Sarah B. Emery, Stanislas Lyonnet, Jeanne Amiel, Muriel Holder, Andrew A. Heggie, Michael J. Bamshad, Deborah A. Nick- erson, Timothy C. Cox, Anne V. Hing, Jeremy A. Horst, and Michael L. Cunningham. A Human Homeotic Transformation Resulting from Mu- tations in PLCB4 and GNAI3 Causes Auriculocondylar Syndrome. The American Journal of Human Genetics, 90(5):907–914, May 2012.

[36] Steven R. Young, Shih-Chieh Chuang, and Robert K. S. Wong. Modu- lation of afterpotentials and firing pattern in guinea pig CA3 neurones by group I metabotropic glutamate receptors. The Journal of Physiology, 554(2):371–385, January 2004.

[37] Vincenza Rita Lo Vasco, Giuseppina Cardinale, and Patrizia Polonia. Deletion of PLCB1 gene in schizophrenia-affected patients. Journal of Cellular and Molecular Medicine, 16(4):844–851, April 2012. [38] Murim Choi, Ute I. Scholl, Peng Yue, Peyman Bj¨orklund,Bixiao Zhao, Carol Nelson-Williams, Weizhen Ji, Yoonsang Cho, Aniruddh Patel, Clara J. Men, Elias Lolis, Max V. Wisgerhof, David S. Geller, Shrikant Mane, Per Hellman, Gunnar Westin, G¨oran Akerstr¨om,Wenhui˚ Wang, Tobias Carling, and Richard P. Lifton. K+ Channel Mutations in Adrenal Aldosterone-Producing Adenomas and Hereditary Hypertension. Science, 331(6018):768–772, February 2011. [39] Martin Poot, Marc J. Eleveld, Ruben van ’t Slot, Hans Kristian Ploos van Amstel, and Ron Hochstenbach. Recurrent copy number changes in men- tally retarded children harbour genes involved in cellular localization and the glutamate receptor complex. European Journal of Human Genetics, 18(1):39–46, January 2010.

19 [40] Kazuo Yamada, Yoshimi Iwayama, Tomoko Toyota, Tetsuo Ohnishi, Hisako Ohba, Motoko Maekawa, and Takeo Yoshikawa. Association study of the KCNJ3 gene as a susceptibility candidate for schizophrenia in the Chinese population. Human Genetics, 131(3):443–451, March 2012. [41] Saber Qanbari, Hubert Pausch, Sandra Jansen, Mehmet Somel, Tim M. Strom, Ruedi Fries, Rasmus Nielsen, and Henner Simianer. Classic Selec- tive Sweeps Revealed by Massive Sequencing in Cattle. PLOS Genetics, 10(2):e1004148, February 2014. [42] E. K. Green, D. Grozeva, I. Jones, L. Jones, G. Kirov, S. Caesar, K. Gordon-Smith, C. Fraser, L. Forty, E. Russell, M. L. Hamshere, V. Moskvina, I. Nikolov, A. Farmer, P. McGuffin, P. A. Holmans, M. J. Owen, M. C. O’Donovan, and N. Craddock. The bipolar disorder risk allele at CACNA1C also confers risk of recurrent major depression and of schizophrenia. Molecular Psychiatry, 15(10):1016–1022, October 2010. [43] Kristin L. Bigos, Venkata S. Mattay, Joseph H. Callicott, Richard E. Straub, Radhakrishna Vakkalanka, Bhaskar Kolachana, Thomas M. Hyde, Barbara K. Lipska, Joel E. Kleinman, and Daniel R. Weinberger. Genetic Variation in CACNA1c Affects Brain Circuitries Related to Mental Illness. Archives of General Psychiatry, 67(9):939–945, September 2010. [44] Martin Tesli, Kristina C. Skatun, Olga Therese Ousdal, Andrew Anand Brown, Christian Thoresen, Ingrid Agartz, Ingrid Melle, Srdjan Djurovic, Jimmy Jensen, and Ole A. Andreassen. CACNA1c Risk Variant and Amygdala Activity in Bipolar Disorder, Schizophrenia and Healthy Con- trols. PLOS ONE, 8(2):e56970, February 2013. [45] P. A. Olson. G-Protein-Coupled Receptor Modulation of Striatal CaV1.3 L-Type Ca2+ Channels Is Dependent on a Shank-Binding Domain. Jour- nal of Neuroscience, 25(5):1050–1062, February 2005. [46] Michelle Day, Zhongfeng Wang, Jun Ding, Xinhai An, Cali A Ingham, Andrew F Shering, David Wokosin, Ema Ilijic, Zhuoxin Sun, Allan R Sampson, Enrico Mugnaini, Ariel Y Deutch, Susan R Sesack, Gordon W Arbuthnott, and D James Surmeier. Selective elimination of glutamatergic synapses on striatopallidal neurons in Parkinson disease models. Nature Neuroscience, 9(2):251–259, February 2006. [47] Elena A. B. Azizan, Hanne Poulsen, Petronel Tuluc, Junhua Zhou, Michael V. Clausen, Andreas Lieb, Carmela Maniero, Sumedha Garg, Elena G. Bochukova, Wanfeng Zhao, Lalarukh Haris Shaikh, Cheryl A. Brighton, Ada E. D. Teo, Anthony P. Davenport, Tanja Dekkers, Bas Tops, Benno K¨usters,Jiri Ceral, Giles S. H. Yeo, Sudeshna Guha Neogi, Ian McFarlane, Nitzan Rosenfeld, Francesco Marass, James Hadfield, Wo- jciech Margas, Kanchan Chaggar, Miroslav Solar, Jaap Deinum, An- nette C. Dolphin, I. Sadaf Farooqi, Joerg Striessnig, Poul Nissen, and

20 Morris J. Brown. Somatic mutations in ATP1A1 and CACNA1D underlie a common subtype of adrenal hypertension. Nature Genetics, 45(9):1055– 1060, September 2013. [48] Mary Ann Raghanti, Melissa K. Edler, Alexa R. Stephenson, Emily L. Munger, Bob Jacobs, Patrick R. Hof, Chet C. Sherwood, Ralph L. Hol- loway, and C. Owen Lovejoy. A neurochemical hypothesis for the ori- gin of hominids. Proceedings of the National Academy of Sciences, page 201719666, January 2018. [49] Fran¸cois Beaubien, Reesha Raja, Timothy E. Kennedy, Alyson E. Fournier, and Jean-Fran¸coisCloutier. Slitrk1 is localized to excitatory synapses and promotes their development. Scientific Reports, 6:27343, June 2016. [50] Martin Kuhlwilm and Cedric Boeckx. Genetic differences between humans and other hominins contribute to the ”human condition”. bioRxiv, April 2018. [51] Jesse F. Abelson, Kenneth Y. Kwan, Brian J. O’Roak, Danielle Y. Baek, Althea A. Stillman, Thomas M. Morgan, Carol A. Mathews, David L. Pauls, Mladen-Roko Raˇsin,Murat Gunel, Nicole R. Davis, A. Gulhan Ercan-Sencicek, Danielle H. Guez, John A. Spertus, James F. Leckman, Leon S. Dure, Roger Kurlan, Harvey S. Singer, Donald L. Gilbert, Anita Farhi, Angeliki Louvi, Richard P. Lifton, Nenad Sestan,ˇ and Matthew W. State. Sequence Variants in SLITRK1 Are Associated with Tourette’s Syndrome. Science, 310(5746):317–320, October 2005. [52] Yun Zhou and Niels Christian Danbolt. GABA and Glutamate Trans- porters in Brain. Frontiers in Endocrinology, 4(165), November 2013. [53] Paul Daniel Arnold, Tricia Sicard, Eliza Burroughs, Margaret A. Richter, and James L. Kennedy. Glutamate Transporter Gene SLC1a1 Associ- ated With Obsessive-compulsive Disorder. Archives of General Psychia- try, 63(7):769–776, July 2006. [54] Rageen Rajendram, Sefi Kronenberg, Christie L. Burton, and Paul D. Arnold. Glutamate Genetics in Obsessive-Compulsive Disorder: A Re- view. Journal of the Canadian Academy of Child and Adolescent Psychi- atry, 26(3):205–213, 2017. [55] P. G. Eusebi, O. Cort´es, C. Carleos, S. Dunner, and J. Ca˜non.Detection of selection signatures for agonistic behaviour in cattle. Journal of Animal Breeding and Genetics = Zeitschrift Fur Tierzuchtung Und Zuchtungsbi- ologie, April 2018. [56] Amanda L. Pendleton, Feichen Shen, Angela M. Taravella, Sarah Emery, Krishna R. Veeramah, Adam R. Boyko, and Jeffrey M. Kidd. Comparison of village dog and wolf genomes highlights the role of the neural crest in dog domestication. BMC Biology, 16:64, June 2018.

21 [57] Toshiki Takenouchi, Noriko Hashida, Chiharu Torii, Rika Kosaki, Takao Takahashi, and Kenjiro Kosaki. 1p34.3 deletion involving GRIK3: Further clinical implication of GRIK family glutamate receptors in the pathogen- esis of developmental delay. American Journal of Medical Genetics. Part A, 164A(2):456–460, February 2014.

[58] S. Begni, M. Popoli, S. Moraschi, S. Bignotti, G. B. Tura, and M. Gennarelli. Association between the ionotropic glutamate receptor kainate 3 (GRIK3 ) ser310ala polymorphism and schizophrenia. Molecu- lar Psychiatry, 7(4):416–418, April 2002. [59] Gary M. Wilson, Stephane Flibotte, Vikramjit Chopra, Brianna L. Mel- nyk, William G. Honer, and Robert A. Holt. DNA copy-number analysis in bipolar disorder and schizophrenia reveals aberrations in genes involved in glutamate signaling. Human Molecular Genetics, 15(5):743–749, March 2006. [60] H. H. Schiffer and S. F. Heinemann. Association of the human kainate receptor GluR7 gene (GRIK3) with recurrent major depressive disorder. American Journal of Medical Genetics Part B: Neuropsychiatric Genetics, 144B(1):20–26, January 2007. [61] A. L. Gray, T. M. Hyde, A. Deep-Soboslay, J. E. Kleinman, and M. S. Sodhi. Sex differences in glutamate receptor gene expression in major depression and suicide. Molecular Psychiatry, 20(9):1057–1068, September 2015. [62] Alexandra Schosser, Amy W. Butler, Rudolf Uher, Mandy Y. Ng, Sarah Cohen-Woods, Nick Craddock, Mike J. Owen, Ania Korszun, Michael Gill, John Rice, Joanna Hauser, Neven Henigsberg, Wolfgang Maier, Ole Mors, Anna Placentino, Marcella Rietschel, Daniel Souery, Martin Preisig, Ian W. Craig, Anne E. Farmer, Cathryn M. Lewis, and Peter McGuffin. Genome-wide association study of co-occurring anxiety in major depres- sion. The World Journal of Biological Psychiatry, 14(8):611–621, Decem- ber 2013.

[63] Uma Vaidyanathan, Stephen M. Malone, Michael B. Miller, Matt McGue, and William G. Iacono. Heritability and molecular genetic basis of acoustic startle eye blink and affectively modulated startle response: A genome- wide association study. Psychophysiology, 51(12):1285–1299, December 2014.

[64] Alessandra Minelli, Catia Scassellati, Cristian Bonvicini, Jorge Perez, and Massimo Gennarelli. An Association of GRIK3 Ser310ala Functional Polymorphism with Personality Traits. Neuropsychobiology, 59(1):28–33, February 2009.

22 [65] Richard H. P Porter, Sharon L Eastwood, and Paul J Harrison. Distribu- tion of kainate receptor subunit mRNAs in human hippocampus, neocor- tex and cerebellum, and bilateral reduction of hippocampal GluR6 and KA2 transcripts in schizophrenia. Brain Research, 751(2):217–231, March 1997. [66] J. Bah, H. Quach, R. P. Ebstein, R. H. Segman, J. Melke, S. Jamain, M. Rietschel, I. Modai, K. Kanas, O. Karni, B. Lerer, D. Gourion, M. O. Krebs, B. Etain, F. Sch¨urhoff,A. Sz¨oke, M. Leboyer, and T. Bourgeron. Maternal transmission disequilibrium of the glutamate receptor GRIK2 in schizophrenia. NeuroReport, 15(12):1987, August 2004. [67] G. Shaltiel, S. Maeng, O. Malkesman, B. Pearson, R. J. Schloesser, T. Tragon, M. Rogawski, M. Gasior, D. Luckenbaugh, G. Chen, and H. K. Manji. Evidence for the involvement of the kainate receptor subunit GluR6 (GRIK2) in mediating behavioral displays related to behavioral symptoms of mania. Molecular Psychiatry, 13(9):858–872, September 2008. [68] Soon Ae Kim, Jin Hee Kim, Mira Park, In Hee Cho, and Hee Jeong Yoo. Family-based association study between GRIK2 polymorphisms and autism spectrum disorders in the Korean trios. Neuroscience Research, 58(3):332–335, July 2007. [69] Gonzalo Laje, Silvia Paddock, Husseini Manji, A. John Rush, Alexan- der F. Wilson, Dennis Charney, and Francis J. McMahon. Genetic Mark- ers of Suicidal Ideation Emerging During Citalopram Treatment of Major Depression. American Journal of Psychiatry, 164(10):1530–1538, October 2007. [70] Ronald S. Petralia, Ya-Xian Wang, and Robert J. Wenthold. Histological and ultrastructural localization of the kainate receptor subunits, KA2 and GluR6/7, in the rat nervous system using selective antipeptide antibodies. Journal of Comparative Neurology, 349(1):85–110, November 1994. [71] Changhai Cui and Mark L. Mayer. Heteromeric Kainate Receptors Formed by the Coassembly of GluR5, GluR6, and GluR7. Journal of Neuroscience, 19(19):8281–8291, October 1999. [72] S Ozawa. Glutamate receptors in the mammalian central nervous system. Progress in Neurobiology, 54(5):581–618, March 1998. [73] Mohammad Mahdi Motazacker, Benjamin Rainer Rost, Tim Hucho, Ma- soud Garshasbi, Kimia Kahrizi, Reinhard Ullmann, Seyedeh Sedigheh Abedini, Sahar Esmaeeli Nieh, Saeid Hosseini Amini, Chandan Goswami, Andreas Tzschach, Lars Riff Jensen, Dietmar Schmitz, Hans Hilger Rop- ers, Hossein Najmabadi, and Andreas Walter Kuss. A Defect in the Ionotropic Glutamate Receptor 6 Gene (GRIK2) Is Associated with Au- tosomal Recessive Mental Retardation. The American Journal of Human Genetics, 81(4):792–798, October 2007.

23 [74] Maria Clara Bonaglia, Roberto Ciccone, Giorgio Gimelli, Stefania Gimelli, Susan Marelli, Joke Verheij, Roberto Giorda, Rita Grasso, Renato Borgatti, Filomena Pagone, Laura Rodr`ıguez, Maria-Luisa Martinez- Frias, Conny van Ravenswaaij, and Orsetta Zuffardi. Detailed pheno- type–genotype study in five patients with 6q16 deletion: nar- rowing the critical region for Prader–Willi-like phenotype. European Jour- nal of Human Genetics, 16(12):1443–1449, December 2008. [75] M. C´ordoba, S. Rodriguez, D. Gonz´alezMor´on,N. Medina, and M.A. Kauffman. Expanding the spectrum of Grik2 mutations: intellectual dis- ability, behavioural disorder, epilepsy and dystonia: Letter to the Editor. Clinical Genetics, 87(3):293–295, March 2015.

[76] Yomayra F. Guzm´an,Keri Ramsey, Jacob R. Stolz, David W. Craig, Mathew J. Huentelman, Vinodh Narayanan, and Geoffrey T. Swanson. A gain-of-function mutation in the GRIK2 gene causes neurodevelopmental deficits. Neurology: Genetics, 3(e129):1–9, January 2017.

[77] Romolo Caniglia, Elena Fabbri, Pavel Hulva, Barbora Cern´aBolf´ıkov´a,ˇ Milena Jindˇrichov´a,Astrid Vik Stronen, Ihor Dykyy, Alessio Camatta, Paolo Carnier, Ettore Randi, and Marco Galaverni. Wolf outside, dog inside? The genomic make-up of the Czechoslovakian Wolfdog. BMC Genomics, 19(533), July 2018.

[78] Irene Brusini, Miguel Carneiro, Chunliang Wang, Carl-Johan Rubin, Hen- rik Ring, Sandra Afonso, Jos´eA. Blanco-Aguiar, Nuno Ferrand, Nima Rafati, Rafael Villafuerte, Orjan¨ Smedby, Peter Damberg, Finn Hallb¨o¨ok, Mats Fredrikson, and Leif Andersson. Changes in brain architecture are consistent with altered fear processing in domestic rabbits. Proceedings of the National Academy of Sciences, 115(28):7380–7385, July 2018.

[79] Shanelle Ko, Ming-Gao Zhao, Hiroki Toyoda, Chang-Shen Qiu, and Min Zhuo. Altered Behavioral Responses to Noxious Stimuli and Fear in Glu- tamate Receptor 5 (GluR5)- or GluR6-Deficient Mice. Journal of Neuro- science, 25(4):977–984, January 2005.

[80] Jeffrey H. Boyd. Exclusion Criteria of DSM-III: A Study of Co-occurrence of Hierarchy-Free Syndromes. Archives of General Psychiatry, 41(10):983– 989, October 1984. [81] Paul Moran and Sheilagh Hodgins. The Correlates of Comorbid Antisocial Personality Disorder in Schizophrenia. Schizophrenia Bulletin, 30(4):791– 802, January 2004. [82] Jan Volavka. Violence in schizophrenia and bipolar disorder. Psychiatria Danubina, 25(1):24–33, March 2013. [83] A. Sariaslan, H. Larsson, and S. Fazel. Genetic and environmental deter- minants of violence risk in psychotic disorders: a multivariate quantitative

24 genetic study of 1.8 million Swedish twins and siblings. Molecular Psychi- atry, 21(9):1251–1256, September 2016. [84] Richard Delorme, Marie-Odile Krebs, Nadia Chabane, Isabelle Roy, Bruno Millet, Marie Christine Mouren-Simeoni, Wolfgang Maier, Thomas Bourg- eron, and Marion Leboyer. Frequency and transmission of glutamate receptors GRIK2 and GRIK3 polymorphisms in patients with obsessive compulsive disorder. NeuroReport, 15(4):699, March 2004. [85] Aline S. Sampaio, Jesen Fagerness, Jacquelyn Crane, Marion Leboyer, Richard Delorme, David L. Pauls, and S. Evelyn Stewart. Associa- tion Between Polymorphisms in GRIK2 Gene and Obsessive-Compulsive Disorder: A Family-Based Study. CNS Neuroscience & Therapeutics, 17(3):141–147, May 2011. [86] James H Meador-Woodruff, Kenneth L Davis, and Vahram Haroutu- nian. Abnormal Kainate Receptor Expression in Prefrontal Cortex in Schizophrenia. Neuropsychopharmacology, 24(5):545–552, May 2001. [87] Hisham M. Ibrahim, Alan J. Hogg, Daniel J. Healy, Vahram Haroutu- nian, Kenneth L. Davis, and James H. Meador-Woodruff. Ionotropic Glu- tamate Receptor Binding and Subunit mRNA Expression in Thalamic Nuclei in Schizophrenia. American Journal of Psychiatry, 157(11):1811– 1823, November 2000. [88] M`onicaGratac`os,Javier Costas, Rafael de Cid, M`onicaBay´es,Juan R. Gonz´alez,Enrique Baca-Garc´ıa,Yolanda de Diego, Fernando Fern´andez- Aranda, Jos´eFern´andez-Piqueras,Miriam Guitart, Roc´ıoMart´ın-Santos, Lourdes Martorell, Jos´eM. Mench´on,Miquel Roca, Jer´onimoS´aiz-Ruiz, Julio Sanju´an,Marta Torrens, Mikel Urretavizcaya, Joaqu´ınValero, Elis- abet Vilella, Xavier Estivill, and Angel´ Carracedo. Identification of new putative susceptibility genes for several psychiatric disorders by associa- tion analysis of regulatory and non-synonymous SNPs of 306 genes in- volved in neurotransmission and neurodevelopment. American Journal of Medical Genetics Part B: Neuropsychiatric Genetics, 150B(6):808–816, September 2009. [89] Niklas Krumm, Tychele N. Turner, Carl Baker, Laura Vives, Kiana Moha- jeri, Kali Witherspoon, Archana Raja, Bradley P. Coe, Holly A. Stessman, Zong-Xiao He, Suzanne M. Leal, Raphael Bernier, and Evan E. Eichler. Excess of rare, inherited truncating mutations in autism. Nature Genetics, 47(6):582–588, June 2015. [90] Xu Wang, Lenore Pipes, Lyudmila N. Trut, Yury Herbeck, Anastasiya V. Vladimirova, Rimma G. Gulevich, Anastasiya V. Kharlamova, Jennifer L. Johnson, Gregory M. Acland, Anna V. Kukekova, and Andrew G. Clark. Genomic responses to selection for tame/aggressive behaviors in the silver fox (Vulpes vulpes). Proceedings of the National Academy of Sciences, page 201800889, September 2018.

25 [91] Adam G. Walker, Cody J. Wenthur, Zixiu Xiang, Jerri M. Rook, Kyle A. Emmitte, Colleen M. Niswender, Craig W. Lindsley, and P. Jeffrey Conn. Metabotropic glutamate receptor 3 activation is required for long-term depression in medial prefrontal cortex and fear extinction. Proceedings of the National Academy of Sciences, 112(4):1196–1201, January 2015. [92] Darryle D. Schoepp, Rebecca A. Wright, Louise R. Levine, Brenda Gay- dos, and William Z. Potter. LY354740, an mGlu2/3 Receptor Agonist as a Novel Approach to Treat Anxiety/Stress. Stress, 6(3):189–197, January 2003. [93] Chad J. Swanson, Mark Bures, Michael P. Johnson, Anni-Maija Linden, James A. Monn, and Darryle D. Schoepp. Metabotropic glutamate re- ceptors as novel targets for anxiety and stress disorders. Nature Reviews Drug Discovery, 4(2):131–144, February 2005. [94] Toshiharu Shimazaki, Michihiko Iijima, and Shigeyuki Chaki. Anxiolytic- like activity of MGS0039, a potent group II metabotropic glutamate re- ceptor antagonist, in a marble-burying behavior test. European Journal of Pharmacology, 501(1):121–125, October 2004. [95] Andrzej Pilc, Shigeyuki Chaki, Gabriel Nowak, and Jeffrey M. Witkin. Mood disorders: Regulation by metabotropic glutamate receptors. Bio- chemical Pharmacology, 75(5):997–1006, March 2008. [96] P. Jeffrey Conn, Craig W. Lindsley, and Carrie K. Jones. Activation of metabotropic glutamate receptors as a novel approach for the treatment of schizophrenia. Trends in pharmacological sciences, 30(1):25–31, January 2009. [97] Jos´eL. Moreno, Stuart C. Sealfon, and Javier Gonz´alez-Maeso.Group II metabotropic glutamate receptors and schizophrenia. Cellular and Molec- ular Life Sciences, 66(23):3777, December 2009. [98] Niamh L. O’Brien, Michael J. Way, Radhika Kandaswamy, Alessia Fiorentino, Sally I. Sharp, Giorgia Quadri, Jarram Alex, Adebayo An- jorin, David Ball, Raquin Cherian, Karim Dar, Aynur Gormez, Irene Guerrini, Mathis Heydtmann, Audrey Hillman, Sudheer Lankappa, Greg Lydall, Aideen O’Kane, Shamir Patel, Digby Quested, Iain Smith, Al- lan D. Thomson, Nicholas J. Bass, Marsha Y. Morgan, David Curtis, and Andrew McQuillin. The functional GRM3 Kozak sequence variant rs148754219 affects the risk of schizophrenia and alcohol dependence as well as bipolar disorder:. Psychiatric Genetics, 24(6):277–278, December 2014. [99] Jillian P. Casey, Tiago Magalhaes, Judith M. Conroy, Regina Regan, Naisha Shah, Richard Anney, Denis C. Shields, Brett S. Abrahams, Joana Almeida, Elena Bacchelli, Anthony J. Bailey, Gillian Baird, Agatino Battaglia, Tom Berney, Nadia Bolshakova, Patrick F. Bolton, Thomas

26 Bourgeron, Sean Brennan, Phil Cali, Catarina Correia, Christina Corsello, Marc Coutanche, Geraldine Dawson, Maretha de Jonge, Richard De- lorme, Eftichia Duketis, Frederico Duque, Annette Estes, Penny Farrar, Bridget A. Fernandez, Susan E. Folstein, Suzanne Foley, Eric Fombonne, Christine M. Freitag, John Gilbert, Christopher Gillberg, Joseph T. Gless- ner, Jonathan Green, Stephen J. Guter, Hakon Hakonarson, Richard Holt, Gillian Hughes, Vanessa Hus, Roberta Igliozzi, Cecilia Kim, Sabine M. Klauck, Alexander Kolevzon, Janine A. Lamb, Marion Leboyer, Ann Le Couteur, Bennett L. Leventhal, Catherine Lord, Sabata C. Lund, Elena Maestrini, Carine Mantoulan, Christian R. Marshall, Helen McConachie, Christopher J. McDougle, Jane McGrath, William M. McMahon, Ali- son Merikangas, Judith Miller, Fiorella Minopoli, Ghazala K. Mirza, Jeff Munson, Stanley F. Nelson, Gudrun Nygren, Guiomar Oliveira, Alis- tair T. Pagnamenta, Katerina Papanikolaou, Jeremy R. Parr, Barbara Parrini, Andrew Pickles, Dalila Pinto, Joseph Piven, David J. Posey, Annemarie Poustka, Fritz Poustka, Jiannis Ragoussis, Bernadette Roge, Michael L. Rutter, Ana F. Sequeira, Latha Soorya, InˆesSousa, Nuala Sykes, Vera Stoppioni, Raffaella Tancredi, Ma¨ıt´eTauber, Ann P. Thomp- son, Susanne Thomson, John Tsiantis, Herman Van Engeland, John B. Vincent, Fred Volkmar, Jacob A. S. Vorstman, Simon Wallace, Kai Wang, Thomas H. Wassink, Kathy White, Kirsty Wing, Kerstin Wittemeyer, Brian L. Yaspan, Lonnie Zwaigenbaum, Catalina Betancur, Joseph D. Buxbaum, Rita M. Cantor, Edwin H. Cook, Hilary Coon, Michael L. Cuccaro, Daniel H. Geschwind, Jonathan L. Haines, Joachim Hallmayer, Anthony P. Monaco, John I. Nurnberger, Margaret A. Pericak-Vance, Ger- ard D. Schellenberg, Stephen W. Scherer, James S. Sutcliffe, Peter Szat- mari, Veronica J. Vieland, Ellen M. Wijsman, Andrew Green, Michael Gill, Louise Gallagher, Astrid Vicente, and Sean Ennis. A novel approach of homozygous haplotype sharing identifies candidate genes in autism spectrum disorder. Human Genetics, 131(4):565–579, April 2012. [100] Yu-Wen Chen, Hui-Ching Lin, Ming-Chong Ng, Ya-Hsin Hsiao, Chao- Chuan Wang, Po-Wu Gean, and Po See Chen. Activation of mGluR2/3 underlies the effects of N-acetylcystein on amygdala-associated autism- like phenotypes in a valproate-induced rat model of autism. Frontiers in Behavioral Neuroscience, 8, 2014. [101] Lena Wischhof and Michael Koch. Pre-treatment with the mGlu2/3 recep- tor agonist LY379268 attenuates DOI-induced impulsive responding and regional c-Fos protein expression. Psychopharmacology, 219(2):387–400, January 2012.

[102] Andrea Benazzo, Emiliano Trucchi, James A. Cahill, Pierpaolo Maisano Delser, Stefano Mona, Matteo Fumagalli, Lynsey Bunnefeld, Luca Cor- netti, Silvia Ghirotto, Matteo Girardi, Lino Ometto, Alex Panziera, Omar Rota-Stabelli, Enrico Zanetti, Alexandros Karamanlidis, Claudio Groff, Ladislav Paule, Leonardo Gentile, Carles Vil`a,Saverio Vicario, Luigi Boi-

27 tani, Ludovic Orlando, Silvia Fuselli, Cristiano Vernesi, Beth Shapiro, Paolo Ciucci, and Giorgio Bertorelle. Survival and divergence in a small group: The extraordinary genomic history of the endangered Apennine brown bear stragglers. Proceedings of the National Academy of Sciences, 114(45):E9589–E9597, November 2017.

[103] Wenjin Li, Kang Ju, Zhiqiang Li, Kuanjun He, Jianhua Chen, Qingzhong Wang, Beimeng Yang, Lin An, Guoyin Feng, Weiming Sun, Juan Zhou, Shasha Zhang, Pingping Song, Raja Amjad Waheed Khan, Weidong Ji, and Yongyong Shi. Significant association of GRM7 and GRM8 genes with schizophrenia and major depressive disorder in the Han Chinese popula- tion. European Neuropsychopharmacology, 26(1):136–146, January 2016.

[104] Jing Li, Huaqing Meng, Wan Cao, and Tian Qiu. MiR-335 is involved in major depression disorder and antidepressant treatment through targeting GRM4. Neuroscience Letters, 606:167–172, October 2015. [105] Tahereh Dadkhah, Simin Rahimi-Aliabadi, Javad Jamshidi, Hamid Ghaedi, Shaghyegh Taghavi, Parasto Shokraeian, Haleh Akhavan-Niaki, Abbas Tafakhori, Mina Ohadi, and Hossein Darvish. A genetic variant in miRNA binding site of glutamate receptor 4, metabotropic (GRM4) is associated with increased risk of major depressive disorder. Journal of Affective Disorders, 208:218–222, January 2017.

[106] Robert M. Duvoisin, Laura Villasana, Timothy Pfankuch, and Jacob Raber. Sex-dependent cognitive phenotype of mice lacking mGluR8. Be- havioural Brain Research, 209(1):21–26, May 2010. [107] Robert M. Duvoisin, Laura Villasana, Matthew J. Davis, Danny G. Winder, and Jacob Raber. Opposing roles of mGluR8 in measures of anxiety involving non-social and social challenges. Behavioural Brain Re- search, 221(1):50–54, August 2011. [108] Richard M. O’Connor, Deepak R. Thakker, Markus Schmutz, Herman van der Putten, Daniel Hoyer, Peter J. Flor, and John F. Cryan. Adult siRNA-induced knockdown of mGlu7 receptors reduces anxiety in the mouse. Neuropharmacology, 72:66–73, September 2013. [109] Miwako Masugi, Mineto Yokoi, Ryuichi Shigemoto, Keiko Muguruma, Yasuyoshi Watanabe, Gilles Sansig, Herman van der Putten, and Shige- tada Nakanishi. Metabotropic Glutamate Receptor Subtype 7 Ablation Causes Deficit in Fear Response and Conditioned Taste Aversion. Journal of Neuroscience, 19(3):955–963, February 1999.

[110] Susanne Schmid and Markus Fendt. Effects of the mGluR8 agonist (S)-3,4-DCPG in the lateral amygdala on acquisition/expression of fear- potentiated startle, synaptic transmission, and plasticity. Neuropharma- cology, 50(2):154–164, February 2006.

28 [111] Hiromi Takaki, Rumiko Kikuta, Hiroki Shibata, Hideaki Ninomiya, Nobu- tada Tashiro, and Yasuyuki Fukumaki. Positive associations of polymor- phisms in the metabotropic glutamate receptor type 8 gene ( GRM8 ) with schizophrenia. American Journal of Medical Genetics, 128B(1):6–14, July 2004.

[112] Tsuyuka Ohtsuki, Minori Koga, Hiroki Ishiguro, Yasue Horiuchi, Makoto Arai, Kazuhiro Niizato, Masanari Itokawa, Toshiya Inada, Nakao Iwata, Shyuji Iritani, Norio Ozaki, Hiroshi Kunugi, Hiroshi Ujike, Yuichiro Watanabe, Toshiuki Someya, and Tadao Arinami. A polymorphism of the metabotropic glutamate receptor mGluR7 (GRM7) gene is associated with schizophrenia. Schizophrenia Research, 101(1):9–16, April 2008.

[113] Clarissa Ganda, Sibylle G. Schwab, Nurmiati Amir, Heriani Heri- ani, Irman Irmansyah, Agung Kusumawardhani, Martina Nasrun, Ika Widyawati, Wolfgang Maier, and Dieter B. Wildenauer. A family-based association study of DNA sequence variants in GRM7 with schizophrenia in an Indonesian population. International Journal of Neuropsychophar- macology, 12(9):1283–1289, October 2009. [114] Subin Park, Sun-Woo Jung, Boong-Nyun Kim, Soo-Churl Cho, Min-Sup Shin, Jae-Won Kim, Hee Jeong Yoo, Dae-Yeon Cho, Un-Sun Chung, Jung- Woo Son, and Hyo-Won Kim. Association between the GRM7 rs3792452 polymorphism and attention deficit hyperacitiveity disorder in a Korean sample. Behavioral and Brain Functions, 9(1):1, January 2013. [115] Josephine Elia, Joseph T. Glessner, Kai Wang, Nagahide Takahashi, Co- rina J. Shtir, Dexter Hadley, Patrick M. A. Sleiman, Haitao Zhang, Cecilia E. Kim, Reid Robison, Gholson J. Lyon, James H. Flory, Jonathan P. Bradfield, Marcin Imielinski, Cuiping Hou, Edward C. Frack- elton, Rosetta M. Chiavacci, Takeshi Sakurai, Cara Rabin, Frank A. Mid- dleton, Kelly A. Thomas, Maria Garris, Frank Mentch, Christine M. Freitag, Hans-Christoph Steinhausen, Alexandre A. Todorov, Andreas Reif, Aribert Rothenberger, Barbara Franke, Eric O. Mick, Herbert Roey- ers, Jan Buitelaar, Klaus-Peter Lesch, Tobias Banaschewski, Richard P. Ebstein, Fernando Mulas, Robert D. Oades, Joseph Sergeant, Edmund Sonuga-Barke, Tobias J. Renner, Marcel Romanos, Jasmin Romanos, An- dreas Warnke, Susanne Walitza, Jobst Meyer, Haukur P´almason,Chris- tiane Seitz, Sandra K. Loo, Susan L. Smalley, Joseph Biederman, Lindsey Kent, Philip Asherson, Richard J. L. Anney, J. William Gaynor, Philip Shaw, Marcella Devoto, Peter S. White, Struan F. A. Grant, Joseph D. Buxbaum, Judith L. Rapoport, Nigel M. Williams, Stanley F. Nelson, Stephen V. Faraone, and Hakon Hakonarson. Genome-wide copy number variation study associates metabotropic glutamate receptor gene networks with attention deficit hyperactivity disorder. Nature Genetics, 44(1):78– 84, January 2012.

29 [116] Yi Liu, Yanqing Zhang, Dongmei Zhao, Rui Dong, Xiaomeng Yang, Kris- tiina Tammimies, Mohammed Uddin, Stephen W. Scherer, and Zhong- tao Gai. Rare de novo deletion of metabotropic glutamate receptor 7 (GRM7) gene in a patient with autism spectrum disorder. American Jour- nal of Medical Genetics Part B: Neuropsychiatric Genetics, 168(4):258– 264, 2015. [117] F. J. Serajee, H. Zhong, R. Nabi, and A. H. M. Mahbubul Huq. The metabotropic glutamate receptor 8 gene at 7q31: partial duplication and possible association with autism. Journal of Medical Genetics, 40(4):e42– e42, April 2003. [118] Radhika Kandaswamy, Andrew McQuillin, David Curtis, and Hugh Gurl- ing. Allelic association, DNA resequencing and copy number variation at the metabotropic glutamate receptor GRM7 gene locus in bipolar dis- order. American Journal of Medical Genetics Part B: Neuropsychiatric Genetics, 165(4):365–372, May 2014. [119] Noriko Sangu, Keiko Shimojima, Yuya Takahashi, Tsukasa Ohashi, Jun Tohyama, and Toshiyuki Yamamoto. A 7q31.33q32.1 microdeletion in- cluding LRRC4 and GRM8 is associated with severe intellectual disabil- ity and characteristics of autism. Variation, 4:17001, February 2017. [120] M. Daniele Fallin, Virginia K. Lasseter, Dimitrios Avramopoulos, Kristin K. Nicodemus, Paula S. Wolyniec, John A. McGrath, Gary Steel, Gerald Nestadt, Kung-Yee Liang, Richard L. Huganir, David Valle, and Ann E. Pulver. Bipolar I Disorder and Schizophrenia: A 440–Single- Nucleotide Polymorphism Screen of 64 Candidate Genes among Ashkenazi Jewish Case-Parent Trios. The American Journal of Human Genetics, 77(6):918–936, December 2005. [121] J´erˆomeAJ Becker, Daniel Clesse, Coralie Spiegelhalter, Yannick Schwab, Julie Le Merrer, and Brigitte L. Kieffer. Autistic-Like Syndrome in Mu Opioid Receptor Null Mice is Relieved by Facilitated mGluR4 Activity. Neuropsychopharmacology, 39(9):2049–2060, August 2014. [122] Matthew J. Davis, Tammie Haley, Robert M. Duvoisin, and Jacob Raber. Measures of anxiety, sensorimotor function, and memory in male and fe- male mGluR4-/- mice. Behavioural Brain Research, 229(1):21–28, April 2012. [123] Sarah N. Isherwood, Trevor W. Robbins, Janet R. Nicholson, Jeffrey W. Dalley, and Anton Pekcec. Selective and interactive effects of D2 receptor antagonism and positive allosteric mGluR4 modulation on waiting impul- sivity. Neuropharmacology, 123:249–260, September 2017. [124] Ahmad Seif Kanaan, Sarah Gerasch, Isabel Garc´ıa-Garc´ıa,Leonie Lampe, Andr´ePampel, Alfred Anwander, Jamie Near, Harald E. M¨oller,and

30 Kirsten M¨uller-Vahl. Pathological glutamatergic neurotransmission in Gilles de la Tourette syndrome. Brain, 140(1):218–234, January 2017. [125] Marco A Grados, Elizabeth B Atkins, Gabriela I Kovacikova, and Erin McVicar. A selective review of glutamate pharmacological therapy in obsessive–compulsive and related disorders. Psychology Research and Be- havior Management, 8:115–131, April 2015. [126] Marion Wittmann, Michael J. Marino, Stefania Risso Bradley, and P. Jef- frey Conn. Activation of Group III mGluRs Inhibits GABAergic and Glu- tamatergic Transmission in the Substantia Nigra Pars Reticulata. Journal of Neurophysiology, 85(5):1960–1968, May 2001.

[127] Antonio Rodr´ıguez-Morenoand Talvinder S. Sihra. Kainate receptors with a metabotropic modus operandi. Trends in Neurosciences, 30(12):630– 637, December 2007. [128] Sergio Valbuena and Juan Lerma. Non-canonical Signaling, the Hidden Life of Ligand-Gated Ion Channels. Neuron, 92(2):316–329, October 2016.

[129] Jos´eV. Negrete-D´ıaz,Talvinder S. Sihra, Gonzalo Flores, and Antonio Rodr´ıguez-Moreno. Non-canonical Mechanisms of Presynaptic Kainate Receptors Controlling Glutamate Release. Frontiers in Molecular Neuro- science, 11, April 2018.

[130] M. P. Mattson, P. Dou, and S. B. Kater. Outgrowth-regulating actions of glutamate in isolated hippocampal pyramidal neurons. Journal of Neuro- science, 8(6):2087–2100, June 1988. [131] Antonio Rodr´ıguez-Moreno and Juan Lerma. Kainate Receptor Mod- ulation of GABA Release Involves a Metabotropic Function. Neuron, 20(6):1211–1218, June 1998. [132] Milos M. Petrovic, Silvia Viana da Silva, James P. Clement, Ladislav Vyklicky, Christophe Mulle, Inmaculada M. Gonz´alez-Gonz´alez, and Jeremy M. Henley. Metabotropic action of postsynaptic kainate recep- tors triggers hippocampal long-term potentiation. Nature Neuroscience, 20(4):529–539, April 2017. [133] S. Bahn, B. Volk, and W. Wisden. Kainate receptor gene expression in the developing rat brain. Journal of Neuroscience, 14(9):5525–5547, September 1994. [134] W Wisden and P H Seeburg. A Complex Mosaic of High-Affinity Kainate Receptors in Rat Brain. Journal of Neuroscience, 13(8):3582–3598, 1993. [135] He Li, Aiqin Chen, Guoqiang Xing, Mei-Ling Wei, and Michael A. Rogawski. Kainate receptor-mediated heterosynaptic facilitation in the amygdala. Nature Neuroscience, 4(6):612–620, June 2001.

31 [136] Steven E. Scherer and Vittorio Gallo. Expression and regulation of kainate and AMPA receptors in the rat neural tube. Journal of Neuroscience Research, 52(3):356–368, May 1998. [137] Laura M Ritter, Alan S Unis, and James H Meador-Woodruff. Ontogeny of ionotropic glutamate receptor expression in human fetal brain. Devel- opmental Brain Research, 127(2):123–133, April 2001. [138] Haesung Lee and Ben H. Choi. Density and distribution of excitatory amino acid receptors in the developing human fetal brain: A quantitative autoradiographic study. Experimental Neurology, 118(3):284–290, Decem- ber 1992. [139] A. Represa, E. Tremblay, and Y. Ben-Ari. Transient increase of NMDA- binding sites in human hippocampus during development. Neuroscience Letters, 99(1):61–66, April 1989. [140] Kenji Sakimura, Hideaki Bujo, Etsuko Kushiya, Kazuaki Araki, Makoto Yamazaki, Masatoshi Yamazaki, Hiroyuki Meguro, Akira Warashina, Shosaku Numa, and Masayoshi Mishina. Functional expression from cloned cDNAs of glutamate receptor species responsive to kainate and quisqualate. FEBS Letters, 272(1):73–80, October 1990. [141] Michael Hollmann and Stephen Heinemann. Cloned Glutamate Receptors. Annual Review of Neuroscience, 17(1):31–108, March 1994. [142] Alfonso Represa, Evelyne Tremblay, Damien Schoevart, and Yehezkel Ben-Ari. Development of high affinity kainate binding sites in human and rat hippocampi. Brain Research, 384(1):170–174, October 1986. [143] Gary W. Mathern, James K. Pretorius, Harley I. Kornblum, Delia Men- doza, Alana Lozada, Joao P. Leite, Leila Chimelli, Donald E. Born, Itzhak Fried, Americo C. Sakamoto, Joao A. Assirati, Warwick J. Pea- cock, George A. Ojemann, and P. David Adelson. Altered Hippocam- pal Kainate-Receptor mRNA Levels in Temporal Lobe Epilepsy Patients. Neurobiology of Disease, 5(3):151–176, September 1998. [144] E. Tremblay, A. Represa, and Y. Ben-Ari. Autoradiographic localization of binding sites in the human hippocampus. Brain research, 343(2):378–382, September 1985. [145] Paul F. Good, George W. Huntley, Scott W. Rogers, Stephen F. Heine- mann, and John H. Morrison. Organization and quantitative analysis of kainate receptor subunit GluR5-7 immunoreactivity in monkey hippocam- pus. Brain Research, 624(1):347–353, October 1993. [146] Francine M. Benes, Mark S. Todtenkopf, and Paul Kostoulakos. GluR5,6,7 subunit immunoreactivity on apical dendrites in hippocam- pus of schizophrenics and manic depressives. Hippocampus, 11(5):482–491, October 2001.

32 [147] R. B. Meeker, R. S. Greenwood, and J. N. Hayward. Glutamate receptors in the rat hypothalamus and pituitary. Endocrinology, 134(2):621–629, February 1994. [148] Jean-Michel Aubry, Viktor Bartanusz, Sonia Pagliusi, Pierre Schulz, and Jozsef Z. Kiss. Expression of ionotropic glutamate receptor subunit mRNAs by paraventricular corticotropin-releasing factor (CRF) neurons. Neuroscience Letters, 205(2):95–98, February 1996. [149] James P. Herman, Ozhan Eyigor, Dana R. Ziegler, and Lothar Jennes. Expression of ionotropic glutamate receptor subunit mRNAs in the hy- pothalamic paraventricular nucleus of the rat. Journal of Comparative Neurology, 422(3):352–362, June 2000. [150] A. N. Van Den Pol, K. Obrietan, V. Cao, and P. Q. Trombley. Embryonic hypothalamic expression of functional glutamate receptors. Neuroscience, 67(2):419–439, July 1995. [151] H. Ohishi, R. Shigemoto, S. Nakanishi, and N. Mizuno. Distribution of the messenger RNA for a metabotropic glutamate receptor, mGluR2, in the central nervous system of the rat. Neuroscience, 53(4):1009–1018, April 1993. [152] H. Ohishi, R. Shigemoto, S. Nakanishi, and N. Mizuno. Distribution of the mRNA for a metabotropic glutamate receptor (mGluR3) in the rat brain: an in situ hybridization study. The Journal of Comparative Neurology, 335(2):252–266, September 1993. [153] Hitoshi Ohishi, Chihiro Akazawa, Ryuichi Shigemoto, Shigetada Nakan- ishi, and Noboru Mizuno. Distributions of the mRNAs for L-2-amino-4- phosphonobutyrate-sensitive metabotropic glutamate receptors, mGluR4 and mGluR7, in the rat brain. Journal of Comparative Neurology, 360(4):555–570, October 1995. [154] Colleen M. Niswender and P. Jeffrey Conn. Metabotropic Glutamate Re- ceptors: Physiology, Pharmacology, and Disease. Annual review of phar- macology and toxicology, 50:295–322, July 2010.

[155] P. J. Flor, K. Lindauer, I. P¨uttner,D. R¨uegg,S. Lukic, T. Kn¨opfel,and R. Kuhn. Molecular Cloning, Functional Expression and Pharmacological Characterization of the Human Metabotropic Glutamate Receptor Type 2. European Journal of Neuroscience, 7(4):622–629, April 1995. [156] PJ Harrison, L. Lyon, LJ Sartorius, Pwj Burnet, and TA Lane. Re- view: The group II metabotropic glutamate receptor 3 (mGluR3, mGlu3, GRM3): expression, function and involvement in schizophrenia. Journal of Psychopharmacology, 22(3):308–322, May 2008.

33 [157] Ingmar Bl¨umcke, Karsten Behle, Barbara Malitschek, Rainer Kuhn, Thomas Kn¨opfel,Helmut K. Wolf, and Otmar D. Wiestler. Immuno- histochemical distribution of metabotropic glutamate receptor subtypes mGluR1b, mGluR2/3, mGluR4a and mGluR5 in human hippocampus. Brain Research, 736(1):217–226, October 1996.

[158] S. R. Bradley, A. I. Levey, S. M. Hersch, and P. J. Conn. Immunocyto- chemical localization of group III metabotropic glutamate receptors in the hippocampus with subtype-specific antibodies. Journal of Neuroscience, 16(6):2044–2056, March 1996. [159] P. J. Flor, S. Lukic, D. R¨uegg,T. Leonhardt, T. Kn¨opfel,and R. K¨uhn. Molecular cloning, functional expression and pharmacological characteri- zation of the human metabotropic glutamate receptor type 4. Neurophar- macology, 34(2):149–155, February 1995. [160] Richard C. Meibach and Allan Siegel. Efferent connections of the hip- pocampal formation in the rat. Brain Research, 124(2):197–224, March 1977. [161] Andrew Makoff, Rosalia Lelchuk, Marcus Oxer, Kathleen Harrington, and Piers Emson. Molecular characterization and localization of hu- man metabotropic glutamate receptor type 4. Molecular Brain Research, 37(1):239–248, April 1996.

[162] Ryuichi Shigemoto, Ayae Kinoshita, Eiki Wada, Sakashi Nomura, Hitoshi Ohishi, Masahiko Takada, Peter J. Flor, Akio Neki, Takaaki Abe, Shige- tada Nakanishi, and Noboru Mizuno. Differential Presynaptic Localization of Metabotropic Glutamate Receptor Subtypes in the Rat Hippocampus. Journal of Neuroscience, 17(19):7503–7522, October 1997.

[163] Andrew Makoff, Charles Pilling, Kathleen Harrington, and Piers Emson. Human metabotropic glutamate receptor type 7: Molecular cloning and mRNA distribution in the CNS. Molecular Brain Research, 40(1):165–170, August 1996. [164] Julie A. Saugstad, J. Mark Kinzie, Michi M. Shinohara, Thomas P. Segerson, and Gary L. Westbrook. Cloning and Expression of Rat Metabotropic Glutamate Receptor 8 Reveals a Distinct Pharmacological Profile. Molecular Pharmacology, 51(1):119–125, January 1997. [165] Corrado Corti, Sophie Restituito, Joseph M. Rimland, Isabelle Brabet, Mauro Corsi, Jean Philippe Pin, and Francesco Ferraguti. Cloning and characterization of alternative mRNA forms for the rat metabotropic glu- tamate receptors mGluR7 and mGluR8. European Journal of Neuro- science, 10(12):3629–3641, December 1998. [166] Pari Malherbe, Claudia Kratzeisen, Kenneth Lundstrom, J. Grayson Richards, Richard L. M Faull, and Vincent Mutel. Cloning and functional

34 expression of alternative spliced variants of the human metabotropic glu- tamate receptor 8. Molecular Brain Research, 67(2):201–210, April 1999. [167] G. Casabona, T. Knopfel, R. Kuhn, F. Gasparini, P. Baumann, M. A. Sortino, A. Copani, and F. Nicolettii. Expression and Coupling to Polyphosphoinositide Hydrolysis of Group I Metabotropic Glutamate Re- ceptors in Early Postnatal and Adult Rat Brain. European Journal of Neuroscience, 9(1):12–17, January 1997. [168] S. Scaccianoce, F. Matrisciano, P. Del Bianco, A. Caricasole, V. Di Giorgi Gerevini, I. Cappuccio, D. Melchiorri, G. Battaglia, and F. Nicoletti. Endogenous activation of group-II metabotropic glutamate receptors inhibits the hypothalamic–pituitary–adrenocortical axis. Neu- ropharmacology, 44(5):555–561, April 2003. [169] Prabhat K Ghosh, Namadev Baskaran, and Anthony N van den Pol. De- velopmentally regulated gene expression of all eight metabotropic gluta- mate receptors in hypothalamic suprachiasmatic and arcuate nuclei – a PCR analysis. Developmental Brain Research, 102(1):1–12, August 1997. [170] M. V. Catania, G. B. Landwehrmeyer, C. M. Testa, D. G. Standaert, J. B. Penney, and A. B. Young. Metabotropic glutamate receptors are differentially regulated during development. Neuroscience, 61(3):481–495, August 1994.

[171] Mar´ıaCristina Defagot, Marcelo J. Villar, and Marta C. Antonelli. Differ- ential Localization of Metabotropic Glutamate Receptors during Postna- tal Development. Developmental Neuroscience, 24(4):272–282, December 2002. [172] Stefania Risso Bradley, Howard D. Rees, Hong Yi, Allan I. Levey, and P. Jeffrey Conn. Distribution and Developmental Regulation of Metabotropic Glutamate Receptor 7a in Rat Brain. Journal of Neuro- chemistry, 71(2):636–645, August 1998.

35