Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Dec 20.
Published in final edited form as: Prog Neuropsychopharmacol Biol Psychiatry. 2024 Aug 2;135:111107. doi: 10.1016/j.pnpbp.2024.111107

Aberrant glutamatergic systems underlying impulsive behaviors: insights from clinical and preclinical research

Justin R Yates 1
PMCID: PMC11409449  NIHMSID: NIHMS2016812  PMID: 39098647

Abstract

Impulsivity is a broad construct that often refers to one of several distinct behaviors and can be measured with self-report questionnaires and behavioral paradigms. Several psychiatric conditions are characterized by one or more forms of impulsive behavior, most notably the impulsive/hyperactive subtype of attention-deficit/hyperactivity disorder (ADHD), mood disorders, and substance use disorders. Monoaminergic neurotransmitters are known to mediate impulsive behaviors and are implicated in various psychiatric conditions. However, growing evidence suggests that glutamate, the major excitatory neurotransmitter of the mammalian brain, regulates important functions that become dysregulated in conditions like ADHD. The purpose of the current review is to discuss clinical and preclinical evidence linking glutamate to separate aspects of impulsivity, specifically motor impulsivity, impulsive choice, and affective impulsivity. Hyperactive glutamatergic activity in the corticostriatal and the cerebro-cerebellar pathways are major determinants of motor impulsivity. Conversely, hypoactive glutamatergic activity in frontal cortical areas and hippocampus and hyperactive glutamatergic activity in anterior cingulate cortex and nucleus accumbens mediate impulsive choice. Affective impulsivity is controlled by similar glutamatergic dysfunction observed for motor impulsivity, except a hyperactive limbic system is also involved. Loss of glutamate homeostasis in prefrontal and nucleus accumbens may contribute to motor impulsivity/affective impulsivity and impulsive choice, respectively. These results are important as they can lead to novel treatments for those with a condition characterized by increased impulsivity that are resistant to conventional treatments.

Keywords: glutamate, motor impulsivity, impulsive choice, affective impulsivity

1. Introduction

Impulsivity is a broad construct that encompasses multiple behaviors and can be assessed with self-report questionnaires such as the Barratt Impulsiveness Scale (BIS-11) (Patton et al., 1995) and the UPPS Scale (Whiteside and Lynam, 2001) and with cognitive-behavioral tasks (these tasks will be described in Section 1.1.). Impulsive behaviors are problematic in certain contexts and are defining features of psychiatric conditions such as attention-deficit/hyperactivity disorder (ADHD) and impulse-control disorders like oppositional defiant disorder, conduct disorder, and intermittent explosive disorder (American Psychiatric Association, 2013). In addition to ADHD and impulse-control disorders, impulsive behaviors are observed in psychiatric conditions like schizophrenia, mood disorders, personality disorders (e.g., borderline personality disorder), substance use disorders, and behavioral addictions (Amlung et al., 2019; Evenden, 1999; Hoptman, 2015; Jentsch et al., 2014; Lee et al., 2019). As a multifaceted construct, the current review will focus on three major forms of impulsivity: motor impulsivity, impulsive choice, and affective impulsivity.

1.1. Major Types of Impulsivity

Motor impulsivity (i.e., impulsive action) is often measured with tasks that require individuals to engage in either action cancellation or action restraint. In stop signal reaction time tasks (SSRTTs), participants are required to inhibit responses they have already initiated when presented with a cue (Logan and Cowan, 1984; Logan et al., 1984; Verbruggen and McLaren, 2018). In go/no-go tasks, individuals are required to either initiate a response (go) or inhibit a response (no-go) when presented with different cues (Newman et al., 1985). In a differential reinforcement of low rates of responding (DRL) task, responses are reinforced only if a response occurs after a designated amount of time. For example, when a DRL-10 s schedule is used, a response is reinforced if the subject waits at least 10 s before emitting a response. A premature response causes the timer to reset (Ferster and Skinner, 1957).

Impulsive choice is primarily conceptualized as the inability to delay gratification; in other words, individuals may forgo a better outcome that is delayed in favor of an immediate, but less preferable, alternative. Delay-discounting tasks are commonly used to measure impulsive choice (Rachlin et al., 1991). Individuals are asked to indicate their preference for a small amount of money made available immediately or a large amount of money delivered after a delay (e.g., $100 now vs. $1000 in 2 months). Steeper discounting indicates higher impulsive choice as individuals are unwilling to wait to earn larger monetary rewards.

Affective impulsivity relates to impulsive behaviors that are dictated by strong emotional responses. One form of affective impulsivity is urgency, in which an individual acts rashly while experiencing a highly negative or a highly positive emotion (Cyders and Smith, 2008). Reactive aggression results from a situation in which the individual feels they have little control; the goal is to neutralize the aversive stimulus (e.g., voluntary manslaughter, lashing out at a supervisor following a negative performance review, etc.). This form of aggression contrasts with proactive aggression, which is premediated, with the aim of achieving an internal or an external goal (e.g., murder, armed robbery, sexual assault, etc.) (see Wrangham, 2018 for a review). Aggression can be directed to oneself in the form of self-injurious behavior, with suicide being a highly concerning form of affective impulsivity (Gvion et al., 2015).

1.2. Animal Models of Impulsivity

To further understand the underlying causes of impulsive behaviors, preclinical research is frequently used. One major area of research centers on the use of animal models of ADHD such as the spontaneously hypertensive rat (SHR) (Sagvolden, 2000) and the Naples High Excitability (NHE) rat (Viggiano et al., 2002). The SHR is an inbred strain derived from the Wistar Kyoto (WKY) rat. Relative to inbred (WKY) and outbred (Wistar, Sprague Dawley) control strains, SHRs show enhanced locomotor activity (Cailhol and Mormède, 1999; Dela Peña et al., 2021; Gungor Aydin and Adiguzel, 2023), impaired attention (Jentsch, 2005; Sagvolden and Xu, 2008), increased impulsive choice (Fox et al., 2008; Fox et al., 2023; Sanabria and Killeen, 2008), and increased motor impulsivity as assessed in a DRL task (Sable et al., 2021). The NHE rat is an outbred strain that has been selectively bred from Sprague Dawley rats since 1976 based on behavioral arousal to novelty. While they display attentional deficits, NHE rats do not display general hyperactivity or impulsive behaviors (Viggiano et al., 2002). As such, discussion of animal models of ADHD will be limited to SHRs in the current review.

Impulsive behaviors can be measured in both SHRs and outbred rodent strains using similar procedures as those used with humans, including the SSRTT (Eagle and Robbins, 2003), go/no-go tasks (Carro-Ciampi and Bignami, 1968; Cole and Michaleski, 1986; Livesey et al., 1980; Sakurai and Sugimoto, 1985), and DRL tasks (Hankosky and Gulley, 2013; Simon et al., 2013; Somkuwar et al., 2016). A popular test of motor impulsivity in rodents is the five-choice serial reaction time task (5CSRTT) (Carli et al., 1983), which is an animal analog of the continuous performance task. While the continuous performance task measures sustained and selective attention, the 5CSRTT measures both attention and motor impulsivity. In the 5CSRTT, rodents must respond in one of several apertures depending on which stimulus light is illuminated. Importantly, rodents must wait a certain amount of time before they respond. Premature responses are used as an index of motor impulsivity. The 5CSRTT can be modified such that the number of active apertures is reduced (1CSRTT, 2CSRTT, etc.; e.g., Abbott et al., 2022; Nord et al., 2019; Tsutsui-Kimura et al., 2009; Ucha et al., 2019).

As with humans, delay-discounting procedures are often used to model impulsive choice (Evenden and Ryan, 1996; Green et al., 2010; Mitchell et al., 2006; Pitts and Febbo, 2004; Rajala et al., 2015; Woolverton et al., 2007). There are several permutations of delay discounting. In most versions of delay discounting, subjects choose between a small, immediate alternative and a large, delayed alternative. The delay to receiving the large magnitude alternative can either increase (Evenden and Ryan, 1996) or decrease (Tanno et al., 2014) within a session or across sessions (Mobini et al., 2002). Alternatively, the delay to receiving the large, delayed alternative can be adjusted depending on prior choice for each alternative (adjusting delay procedures) (Mazur, 1987; Perry et al., 2005). Discussing each version of delay discounting in detail is beyond the scope of this review (see Madden and Johnson, 2010; Yates, 2018 for discussions on the various delay discounting procedures).

1.3. Purpose of Review

As a transdiagnostic trait, there is a large body of literature devoted to better understanding the neurobehavioral mechanisms that control impulsivity. From a neurobiological perspective, considerable attention is directed toward monoaminergic neurotransmitter systems in impulsive behaviors in both humans and animals (Cardinal, 2006; Dalley and Ersche, 2019; Evenden, 1999; Winstanley, 2011). However, there is growing interest in understanding glutamatergic control of several psychiatric disorders, including ADHD (Lesch et al., 2013), schizophrenia (Moghaddam and Javitt, 2012), mood disorders (Jun et al., 2014), and addiction (Kalivas, 2009; Pettorruso et al., 2014). As such, the purpose of the current review is to discuss the contributions of glutamate to impulsive behaviors using clinical and preclinical evidence. Before discussing glutamatergic underpinnings of impulsivity, a brief overview of the glutamatergic system is needed.

2. Overview of Glutamatergic System

Glutamate is the major excitatory neurotransmitter in the mammalian nervous system and is critically involved in long-term potentiation (LTP) and long-term depression (LTD), important cellular mechanisms by which learning occurs (Gladding et al., 2009; Lüscher and Malenka, 2012). Primarily synthesized from glial-released glutamine by the mitochondrial enzyme glutaminase (Torgner and Kvamme, 1990), glutamate is packaged in synaptic vesicles by the vesicular glutamate transporter (Eriksen et al., 2020). Release of glutamate into the synapse is mediated by Ca++ influx (Nicholls and Sihra, 1986). Once released into the synapse, glutamate binds to either metabotropic receptors (mGluRs) or ionotropic receptors, with each receptor type being further divided (see Fig. 1 for a schematic of a glutamatergic synapse).

Figure 1.

Figure 1.

A schematic of a glutamatergic synapse. The inset depicts the postsynaptic density. Created with BioRender.com.

There are three groups of mGluRs. Group I mGluRs (mGluR1 and mGluR5) are near-exclusively located on postsynaptic neurons and are Gq-coupled, stimulating the activation of phospholipase Cβ. Group II (mGluR2 and mGluR3) and Group III (mGluR4, mGluR6, mGluR7, and mGluR8) are Gi-coupled, leading to decreased adenylyl cyclase activation. While Group II mGluRs are found on both presynaptic and postsynaptic neurons, Group III mGluRs are predominately isolated to presynaptic terminal buttons (see Niswender and Conn, 2010 for a comprehensive review of mGluRs).

Ionotropic receptors are divided into kainate, AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid) and NMDA (N-methyl-D-aspartate) receptors, although kainate receptors are often grouped with AMPA receptors as non-NMDA receptors (e.g., Agrawal and Fehlings, 1997; Scott et al., 2019). Each ionotropic receptor is composed of four subunits surrounding an ion channel. Instead of activating a G protein, stimulation of kainate/AMPA and NMDA receptors allows positively charged ions to pass through the channel and into the neuron (Na+ for kainate/AMPA; Ca++ and Na+ for NMDA). AMPA receptors are composed of combinations of GluA1, GluA2, GluA3, and GluA4 subunits while kainate receptors contain GluR5, GluR6, GluR7, KA1, and KA2 subunits. NMDA receptors are composed of two GluN1 subunits, and they contain combinations of GluN2A, GluN2B, GluN2C, and GluN2D subunits (see Bettler and Mulle, 1995; Gan et al., 2015; Regan et al., 2015 for discussions on ionotropic receptor structure). At resting membrane potential, a Mg2+ ion blocks the channel of the NMDA receptor. Depolarization of the neuron via neighboring AMPA receptors causes displacement of the Mg2+ ion from the NMDA receptor channel. The NMDA receptor requires two co-agonists: glycine, which binds to the GluN1 subunit, and glutamate, which binds to the GluN2 subunit.

LTP is commonly associated with ionotropic receptors, but mGluRs are also important for this form of synaptic plasticity to occur (Anwyl, 2009). For simplicity, I will focus on NMDA receptor-mediated LTP. In early phase LTP, high-frequency stimulation of glutamatergic neurons leads to Ca++ influx into the postsynaptic neuron, which causes phosphorylation of additional AMPA receptors by protein kinases such as calcium/calmodulin-dependent protein kinase II (CaMKII); this, in turn, increases the postsynaptic neuron’s sensitivity to glutamate. In late phase LTP, increased Ca++ levels lead to protein synthesis and gene transcription via the activation of other protein kinases like protein kinase A (Bliss and Collingridge, 1993; Madison et al., 1991; Malenka and Bear, 2004). LTD is also mediated by ionotropic glutamate receptors. Low-frequency stimulation of the postsynaptic neuron does not sufficiently displace the Mg++ from the channel. Lower levels of Ca++ enter the neuron, leading to activation of different signaling pathways that result in downregulation of AMPA receptors (Malenka and Bear, 2004). Low frequency stimulation of postsynaptic mGluRs leads to LTD as well (Lüscher and Huber, 2010).

Ionotropic glutamate receptors and signaling molecules like CAMKII are constituents of the postsynaptic density (PSD), a large collection of proteins located at the postsynaptic membrane of dendrites of excitatory neurons (see inset of Fig. 1). Cell adhesion molecules, such as neural cadherins and neuroligins, are a critical component of the PSD as they contribute to the formation of synapses (Togashi et al., 2009). In addition to cell adhesion molecules, there are multiple scaffolding proteins that are involved in the formation and functional integrity of glutamatergic synapses, including SH3 and multiple ankyrin repeat domains protein (SHANK) (Monteiro and Feng, 2017), Homer proteins (Shiraishi-Yamaguchi and Furuichi, 2007), membrane-associated guanylate kinases (MAGUKs) like PSD-95 (Olsen and Bredt, 2003), and guanylate kinase-associated protein (GKAP) (Kim et al., 1997) (see Kaizuka and Takumi (2018) for a comprehensive discussion of the PSD). At high concentrations, the GTPase activator synaptic Ras GTPase-activating protein 1 (SynGAP) acts as a scaffolding protein (Zeng et al., 2019). During LTP, the PSD is enlarged, and actin protein expression increases, leading to increased dendritic spine growth. This growth allows for the accommodation of increased AMPA receptor expression (Herring and Nicoll, 2016).

Because excessive glutamate levels lead to excitotoxicity (Olney, 1986), extracellular glutamate is tightly regulated. First, membrane-bound transporters, most commonly the excitatory amino acid transporter-2 (EAAT-2) in humans (Divito and Underhill, 2014) and the glutamate transporter-1 (GLT-1) in rodents (Rimmele and Rosenberg, 2016), transfer excess glutamate from the synapse to neighboring astrocytes. Glutamate can then be metabolized by amidation back to glutamine via glutamine synthetase (Schousboe et al., 2014). Second, the cysteine/glutamate antiporter system (xC) is crucial for maintaining glutamate homeostasis (Warr et al., 1999). The xC system exchanges extracellular cystine for intracellular glutamate located in astrocytes; this exchange allows glutamate to bind to presynaptic Group II/III mGluRs, inhibiting the additional release of glutamate from the presynaptic neuron (Bridges et al., 2012).

Given the importance of glutamate to learning and memory, both mGluRs and ionotropic glutamate receptors are widely located in brain regions associated with these processes, including cerebral cortex and hippocampus (Abe et al., 1992; Blackstone et al., 1992; Catania et al., 1994; Monaghan et al., 1984; Tanabe et al., 1992; see Ozawa et al., 1998 for a review). Many glutamatergic neurons originate from the cerebral cortex and send projections to neighboring cortical glutamatergic neurons, as well as to the brainstem, the striatum, and the thalamus, with the latter structure projecting glutamatergic neurons back to the cortex (Schwartz et al., 2012). Glutamatergic pathways will be discussed in more detail in Section 3.5.

3. Glutamatergic Dysfunction Underlying Impulsive Behaviors

There are several methods that can be used to identify glutamatergic dysfunction underlying impulsive behaviors. Genetic association studies are commonly used to determine if certain groups of individuals are more likely to have a specific allele or a haplotype of a gene compared to the general population. See Table 1 for glutamatergic-related genes and their corresponding proteins. Researchers can also quantify glutamate levels using techniques like magnetic resonance spectroscopy (MRS) (common in human studies) or in vivo microdialysis and voltammetry (used in preclinical studies). To increase accuracy in MRS studies, glutamate levels are often measured in conjunction with glutamine (glutamate-glutamine or Glx); additionally, other excitatory neurotransmitters like aspartate can be quantified in addition to glutamate/Glx. Another way to identify glutamatergic aberrations is to compare mRNA or protein expression between individuals that display high impulsivity to those that exhibit low impulsivity. In behavioral pharmacology experiments, subjects are treated with a drug that targets a receptor, a transporter, or the xC system. These drugs can be delivered systemically or can be infused into the lateral ventricles or into a specific brain region. Behavioral pharmacology approaches can be used in conjunction with neuroscience techniques, allowing for better determination of how glutamate mediates impulsive behaviors.

Table 1.

Genes related to glutamatergic system in humans

Gene Chromosomal Location Corresponding Protein/Enzyme Function
GRM1 6q24.3 mGluR1 Excitatory metabotropic receptor
GRM2 3p21.2 mGluR2 Inhibitory metabotropic receptor
GRM3 7q21.11-q21.12 mGluR3 Inhibitory metabotropic receptor
GRM4 6p21.31 mGluR4 Inhibitory metabotropic receptor
GRM5 11q14.2-q14.3 mGluR5 Excitatory metabotropic receptor
GRM6 5q35.3 mGluR6 Inhibitory metabotropic receptor
GRM7 3p26.1 mGluR7 Inhibitory metabotropic receptor
GRM8 7q31.33 mGluR8 Inhibitory metabotropic receptor
GRIA1 5q33.2 AMPA subunit GluR1 Subunits of AMPA receptor
GRIA2 4q32.1 AMPA subunit GluR2
GRIA3 Xq25 AMPA subunit GluR3
GRIA4 11q22.3 AMPA subunit GluR4
CACNG2 22q12.3 Calcium voltage-gated channel auxiliary subunit gamma 2 Regulates trafficking and channel gating of AMPA receptors
CACNG3 16p12.1 Calcium voltage-gated channel auxiliary subunit gamma 3
CACNG4 17q24.2 Calcium voltage-gated channel auxiliary subunit gamma 4
CACNG5 17q24.2 Calcium voltage-gated channel auxiliary subunit gamma 5
CACNG7 19q13.42 Calcium voltage-gated channel auxiliary subunit gamma 7
CACNG8 19q13.42 Calcium voltage-gated channel auxiliary subunit gamma 8
GRIN1 9q34.3 NMDA subunit GluN1 Binding site of glycine on NMDA receptor
GRIN2A 16p13.2 NMDA subunit GluN2A Binding site of glutamate on NMDA receptor
GRIN2B 12p13.1 NMDA subunit GluN2B
GRIN2C 17q25.1 NMDA subunit GluN2C
GRIN2D 19q13.33 NMDA subunit GluN2D
GRIK1 21q21.3 Kainate subunit 1 Subunits of kainate receptor
GRIK2 6q16.3 Kainate subunit 2
GRIK3 1p34.3 Kainate subunit 3
GRIK4 11q23.3 Kainate subunit 4
GRIK5 19q13.2 Kainate subunit 5
SLC17A7 19q13.33 Vesicular glutamate transporter 1 Package intracellular glutamate into vesicles
SLC17A6 11p14.3 Vesicular glutamate transporter 2
SLC17A8 12q23.1 Vesicular glutamate transporter 3
SLC1A3 5p13.2 Excitatory amino acid transporter 1 Reuptake of extracellular glutamate
SLC1A2 11p13 Excitatory amino acid transporter 2
SLC1A1 9p24.2 Excitatory amino acid transporter 3
SLC1A6 19p13.12 Excitatory amino acid transporter 4
SLC1A7 1p32.3 Excitatory amino acid transporter 5
SLC7A11 4q28.3 Cystine-glutamate antiporter Exchanges cystine for glutamate
GLS 2q32.2 Glutaminase Synthesizes glutamate from glutamine
GLUL 1q25.3 Glutamine synthetase Catalyzes synthesis of glutamine from glutamate
GAD1 2q31.1 Glutamic acid decarboxylase Converts glutamate to gamma-amino butyric acid (GABA)
SHANK1 19q13.33 SHANK1
SHANK2 11q13.3-q13.4 SHANK2
SHANK3 22q13.33 SHANK3
HOMER1 5q14.1 HOMER1
HOMER2 15q25.2 HOMER2
HOMER3 19p13.11 HOMER2
DLG1 3q29 Discs large membrane-associated guanylate kinase (MAGUK) scaffold protein 1 Scaffolding proteins
DLG2 11q14.1 Discs large MAGUK scaffold protein 2
DLG3 Xq13.1 Discs large MAGUK scaffold protein 3
DLG4 17p13.1 Discs large MAGUK scaffold protein 4
DLG5 10q22.3 Discs large MAGUK scaffold protein 5
DLGAP1 18p11.31 DLG associated protein 1
DLGAP2 8p23.3 DLG associated protein 2
DLGAP3 1p34.3 DLG associated protein 3
DLGAP4 20q11.23 DLG associated protein 4
SYNGAP1 6p21.32 Synaptic Ras GTPase activating protein 1

To date, few studies have determined if the glutamatergic system is altered in individuals diagnosed with an impulse-control disorder as defined by the American Psychiatric Association (e.g., oppositional defiant disorder). Although no differences in glutamate or glutamine levels are observed between children and adolescents diagnosed with intermittent explosive disorder relative to controls (Davanzo et al., 2003), more recent evidence suggests that individuals with the C/C genotype of the GRM8 gene are more likely to have conduct disorder compared to individuals with the C/T or the T/T genotypes (Bauer and Covault, 2020). Due to a lack of studies examining individuals with an impulse-control disorder, the current review will focus on other psychiatric conditions characterized, at least partially, by impulsive behaviors, primarily ADHD, mood disorders, and addiction.

3.1. Glutamatergic Genes Implicated in Disorders Characterized by Impulsive Behaviors

The GRIN2B gene, which encodes for the GluN2B subunit of the NMDA receptor, is linked to both ADHD and bipolar disorder (Dorval et al., 2007; Kim et al., 2018; Zhao et al., 2011), and long-term use of alcohol or cocaine leads to upregulated GRIN2B expression in the brain (Enoch et al., 2014). Specifically, the T-C-C haplotype of the GRIN2B gene is transmitted more frequently in individuals with bipolar disorder (Martucci et al., 2006). However, two GRIN2B single nucleotide polymorphisms (SNPs) (rs2300256 and rs2284411) are associated with inattentive symptoms as reported by one’s parent and/or teacher (Dorval et al., 2007; but see Park et al., 2013), while one additional SNP of the GRIN2B gene (rs2284407) is only marginally associated with hyperactive/impulsive symptoms as reported by parents (Dorval et al., 2007). These results suggest that the GluN2B subunit may be more involved in attentional deficits as opposed to impulsive behavior in individuals with ADHD. In addition to the GRIN2B gene, SNPs of ionotropic glutamate receptors are associated with psychiatric disorders characterized, at least in part, by impulsive behaviors. The GRIN2A gene is associated with ADHD (Turic et al., 2004; but see Adams et al., 2004) and heroin use disorder, at least in in African Americans (Levran et al., 2009). SNPs for the CACNG8 gene are linked to ADHD (Bai et al., 2022) and antisocial personality disorder (Peng et al., 2021), and genes encoding kainate receptors are associated with ADHD (Chatterjee et al., 2022) and aggressive behaviors in individuals with bipolar disorder (Ma et al., 2019). Overall, modifications to excitatory receptors seem to serve as a risk factor for the emergence of maladaptive impulsive behaviors.

Genetic association studies also implicate mGluRs in conditions like ADHD and mood disorders. Specifically, SNPs for GRM7 (Noroozi et al., 2019; Zhang et al., 2021a; but see Akutagava-Martins et al., 2014), as well as deletions of copy variants of genes encoding for mGluR1, mGluR5, mGluR7, and mGluR8, are associated with ADHD (Glessner et al., 2023; Liu et al., 2021), whereas SNPs for the GRM4 gene are protective against ADHD (Zhang et al., 2021a). Like ADHD, two SNPs for the GRM7 gene are observed more frequently in individuals with MDD and bipolar II disorder (Noroozi et al., 2019), and copy number variants of two additional GRM7 SNPs are linked to bipolar disorder (Kandaswamy et al., 2014).

Although the results summarized above implicate glutamatergic genes to psychiatric conditions related to impulsive behaviors, one limitation of genetic association studies is that they do not elucidate how glutamatergic dysfunction mediates such behavior. Relatedly, differential gene expression does not guarantee that protein expression and/or function will be altered. Due to this limitation, studies measuring glutamate/Glx levels, as well as mRNA/protein expression, can provide valuable information concerning aberrant glutamatergic systems in individuals with a psychiatric disorder linked to impulsivity. Below, I will provide evidence of glutamatergic alterations observed in individuals with a psychiatric disorder characterized by impulsive behaviors. I will begin by discussing glutamatergic alterations in the peripheral nervous system.

3.2. Glutamatergic Alterations in Peripheral Nervous System

Peripheral nervous system glutamate/Glx levels and mRNA expression can be quantified from one’s blood plasma. Blood plasma glutamate levels are elevated in children with ADHD, individuals with MDD, and individuals with substance use disorders, with corresponding decreased glutamine levels observed in these conditions (Inoshita et al., 2018; Nishi et al., 2015; Ogawa et al., 2018; Sari et al., 2020; Skalny et al., 2021; Umehara et al., 2017; Wang et al., 2021; Wei et al., 2020). Additionally, peripheral glutamate receptors are more sensitive to glutamate stimulation in individuals with MDD (Berk et al., 2001). One potential explanation for increased peripheral glutamate observed in these individuals is deficient activity of the enzyme glutamine synthetase, leading to overexcitation of glutamatergic receptors. However, this is speculative as glutamine synthetase activity in peripheral nervous system has not been directly measured in individuals with impulse-control related disorders. Another possibility is deficient activity of glutamate decarboxylase (GAD), the enzyme responsible for converting glutamate into the major inhibitory neurotransmitter gamma amino-butyric acid (GABA). There is some evidence to support this hypothesis; individuals with the rs3749034 G allele or the rs11542313 C allele of the GAD1 gene score higher in measures of hyperactivity and impulsivity (Bruxel et al., 2016), and decreased peripheral GAD1 mRNA expression is observed in individuals with MDD (Lin et al., 2019). In addition to deficient GAD enzyme activity, decreased functionality of EAATs may contribute to increased peripheral glutamate levels as the SLC1A3 gene is implicated in ADHD (Laurin et al., 2008; van Amen-Hellebrekers et al., 2016).

In contrast to ADHD, MDD, and substance use disorders, glutamine serum levels are elevated in individuals with bipolar disorder (Pålsson et al., 2015), and glutamate levels are lower in individuals with bipolar disorder experiencing their first psychotic episode (Palomino et al., 2007). Lower glutamate levels are also observed in men with internet gaming disorder (Paik et al., 2018). Individuals experiencing acute mania have elevated antibody levels for the GluN2 subunit of the NMDA receptor (Dickerson et al., 2012), perhaps reflecting a compensatory upregulation in response to decreased glutamate serum levels. The decreased glutamate levels may stem from impaired glutaminase activity in peripheral nervous system and may reflect an important mechanism that distinguishes MDD from bipolar disorder and substance use disorders from behavioral addictions. Research is needed to directly test this hypothesis.

The altered glutamatergic activity observed in individuals with ADHD and mood disorders does not, on its own, provide an explanation for increased impulsive behaviors associated with these conditions. Glutamate is an important mediator of the somatic nervous system, as sensory neurons that transmit information regarding nociception, vision, audition, olfaction, and taste rely on glutamatergic signaling (Kalloniatis et al., 1996; Miller et al., 2011; Murphy et al., 2004; Vandenbeuch and Kinnamon, 2016; Weisz et al., 2021). However, glutamatergic control of motor neurons is primarily localized to the central nervous system, with acetylcholine innervating skeletal muscles in the peripheral nervous system (Stifani, 2014). As such, a discussion of glutamatergic disturbances in the central nervous system will be presented next.

3.3. Glutamatergic Alterations in Cerebrospinal Fluid

One way to measure glutamatergic activity in the central nervous system is to quantify glutamate/Glx levels in cerebrospinal fluid (CSF). Because this approach does not elucidate where in the brain aberrant glutamatergic neurotransmission occurs, relatively few studies focus exclusively on measuring Glx concentrations in CSF. Indeed, examination of Glx levels in CSF has largely been confined to individuals with mood disorders. To this end, no differences in Glx levels are observed in patients with MDD or bipolar disorder compared to controls (Garakani et al., 2013; Hashimoto et al., 2016; Pålsson et al., 2015). Yet, the glutamine-to-glutamate ratio is higher in individuals with MDD, suggesting an abnormal glutamine-glutamate cycle (Hashimoto et al., 2016). This finding is interesting as it suggests the opposite relationship between glutamine and glutamate levels in CSF (increased glutamine and decreased glutamate) compared to the peripheral nervous system (decreased glutamine and increased glutamate). Additional research shows increased glutamine levels in CSF of individuals with MDD (Levine et al., 2000), and individuals with refractory depressive disorders (i.e., those with MDD or bipolar disorder that do not respond to various pharmacotherapies) have lower glutamate levels (Frye et al., 2007a).

Although CSF glutamate levels are lower in individuals with depression, glutamate levels are positively correlated with reactive aggression and suicidal ideation in this population (Coccaro et al., 2013; Garakani et al., 2013), indicating that increased glutamate neurotransmission can increase impulsive-like behaviors in those diagnosed with a mood disorder. Additionally, quinolinic acid, an NMDA receptor agonist, is elevated in individuals that have previously attempted suicide, with quinolinic acid levels positively correlating with suicidal ideation (Erhardt et al., 2013). The positive association observed between CSF glutamate levels and aggression is consistent with increased blood glutamate in individuals with antisocial personality disorder (Gulsun et al., 2016). Preclinical research supports this hypothesis as GAD-deficient mice and GLUT-3 knockout mice are more hyperactive compared to controls (Shin et al., 2021; Smith, 2018).

Like mood disorders, decreased CSF glutamate levels are observed in individuals classified as heavy smokers, with these individuals scoring higher in motor impulsivity as assessed with the BIS-11 (Li et al., 2018). One concerning finding is that alcohol withdrawal leads to an increase in CSF glutamate concentrations (Tsai et al., 1998), as well as in peripheral glutamate concentrations (Aliyev et al., 1994). Preclinical models show increased impulsive behaviors during alcohol withdrawal (Irimia et al., 2014; Irimia et al., 2015; Irimia et al., 2017), providing additional support that aberrant glutamatergic transmission exacerbates impulsivity.

The discrepancy in glutamine/glutamate ratios in peripheral and central nervous systems for individuals with a condition like ADHD or MDD may result from a weakened blood-brain barrier. A dysfunctional blood-brain barrier is observed in individuals with MDD (Wu et al., 2022), and neuroinflammation, which can weaken the blood-brain barrier (Galea, 2021), is proposed to act as a risk factor for ADHD (Dunn et al., 2019). Glutamate released in the central nervous system may “leak” through a weakened blood-brain barrier into the peripheral nervous system. The net result would be elevated glutamate levels in peripheral nervous system and lower glutamate levels in CSF. Tangentially related to this point, increased glutamate consumption decreases the odds of being diagnosed with ADHD during childhood whereas increased consumption of aspartate and glycine increases the odds of being diagnosed with ADHD (Holton et al., 2019). By consuming foods high in glutamate, at-risk children for ADHD may be able to replenish glutamate concentrations in the central nervous system, thus preventing the expression of ADHD-associated impulsive behaviors. Interestingly, the significant association between amino acid consumption and ADHD is specific to children/young adolescents, as this association disappears in college-aged individuals (Holton et al., 2019). Children at risk of ADHD may have a weakened blood-brain barrier that becomes stronger over time. This may provide an explanation as to why ADHD is primarily first diagnosed during childhood, with ADHD diagnoses decreasing across the lifespan (Vos and Hartman, 2022).

3.4. Glutamatergic Control of Impulsive Behaviors: Evidence from Behavioral Pharmacology Experiments

Instead of correlating CSF glutamate levels to impulsive behaviors, behavioral pharmacology experiments are used to determine how administration of a glutamatergic ligand affects impulsivity. Given that elevated CSF glutamate levels are associated with affective impulsivity and drug withdrawal-induced impulsivity (see section above), decreasing excitatory neurotransmission should be effective in attenuating impulsive behaviors. To this end, anticonvulsant medications directly inhibit excitatory neurotransmission by either blocking sodium channels or by enhancing GABA function (Stafstrom, 2010; Zanatta et al., 2019), leading to decreased glutamate concentrations (Soeiro-de-Souza et al., 2018) with corresponding increased glutamine levels (Soeiro-de-Souza et al., 2015). Specific to impulsive behavior, anticonvulsants, particularly diavproex (valproate), reduce impulsive aggression in individuals with or at risk for bipolar disorder (Barzman et al., 2005; Barzman et al., 2006; Chang et al., 2006; MacMillan et al., 2006; Saxena et al., 2006), as well as in children with ADHD (Blader et al., 2009) and in individuals with borderline personality disorder (Hollander et al., 2005; Kavoussi and Coccaro, 1998; Tritt et al., 2005). Similarly, the anticonvulsant lamotrigine decreases aggressive behavior in rats (Kumar et al., 2016).

The results described above support the hypothesis that increased glutamatergic transmission leads to increased impulsivity. To better understand the molecular underpinnings of glutamatergic-mediated impulsivity, preclinical behavioral pharmacology experiments and clinical trials can use ligands that selectively bind to a molecular target. These include ionotropic glutamate receptor antagonists, Group I mGluR antagonists, Group II mGluR agonists, and allosteric modulators of glutamate receptors. Allosteric modulators bind to a different site on the receptor compared to agonists and change the receptor’s response to agonists. Positive allosteric modulators increase the affinity and/or efficacy of an agonist while negative allosteric modulators decrease the affinity and/or efficacy of an agonist (see Conn et al., 2009 for a general review of allosteric modulation; see Gasparini and Spooren, 2007 for a review of mGluR allosteric modulators; see Brogi et al., 2019 for a review of ionotropic glutamate receptor allosteric modulators).

Concerning ionotropic glutamate receptors, few studies focus on the effects of AMPA receptor ligands. While the AMPA receptor antagonist CNQX fails to alter impulsive choice (Yates et al., 2015), an AMPA receptor positive allosteric modulator reduces motor impulsivity in the 1CSRTT (Davis-Reyes et al., 2021), which is consistent with the reduced impulsivity observed in individuals with ADHD following treatment with an AMPA receptor positive allosteric modulator (Adler et al., 2012). In contrast to the AMPA receptor, studies examining the effects of NMDA receptor ligands on impulsive behaviors are more common. Antagonists of NMDA receptors can either (a) block the channel opening (i.e., uncompetitive receptor antagonist), (b) bind to the same site as the agonist (i.e., competitive receptor antagonist), or (c) target NMDA receptors that contain a specific subunit (primarily GluN2B-selective antagonists). Some commonly known drugs act as NMDA receptor channel blockers, including ketamine, memantine, and phencyclidine. One seemingly paradoxical finding is that NMDA receptor antagonists increase glutamate concentrations throughout the brain (Abdallah et al., 2018; Amitai et al., 2012; Ito et al., 2006; Léna et al., 2007; Lisek et al., 2017; Moghaddam et al., 1997; Razoux et al., 2007; Zuo et al., 2006). The increased glutamatergic activity following NMDA receptor antagonism is most likely due the inhibition of inhibitory interneurons (e.g., Hudson et al., 2020; see Lee and Zhou, 2019). As such, NMDA receptor antagonists, including GluN2B-selective antagonists, frequently increase locomotor hyperactivity (Fredriksson and Archer, 2003; Higgins et al., 2016; Popik et al., 2015) and increase motor impulsivity in outbred rodents across a variety of tasks (Burton and Fletcher, 2012; Carli et al., 2011; Higgins et al., 2016; Jentsch and Anzivino, 2004; Terry Jr. et al., 2012; see Table 2 for citations for specific tasks).

Table 2.

Summary of Results of Preclinical Behavioral Pharmacology Experiments Examining Contributions of Glutamate to Motor Impulsivity and Impulsive Choice

Molecular Target Agonist/PAM Antagonist/NAM Region (if not systemically administered) Motor Impulsivity (Specific task) Impulsive Choice References
mGluR 1
JNJ16567083 (EMQMCM) ↓ (DRL) Sukhotina et al. (2008)
JNJ16259685 Yates et al. (2017b)
mGluR 5
RO4917523 ↓ (5CSRTT) --- (5CSRTT) Isherwood et al. (2015)
MTEP NS (5CSRTT) --- (5CSRTT) Isherwood et al. (2015)
MPEP ↓ (5CSRTT) Semenova & Markou (2007)
--- Yates et al. (2017b)
ADX47273 ↓ (5CSRTT) Liu et al. (2008)
--- (5CSRTT) Isherwood et al. (2015)
mGluR 2/3
LY341495 --- (5CSRTT) Semenova & Markou (2007)
LY379268 ↓ (5CSRTT) van der Veen et al. (2021)
mGluR 2
TASP0433864 ↓ (5CSRTT) van der Veen et al. (2021)
mGluR 4
Cpd11 ↑ (Go/no-goa) Piszczek et al. (2022)
↑ (5CSRTT) Isherwood et al. (2017)
AMPA Receptor
HJC0122 ↓ (1CSRTT) Davis-Reyes et al. (2021)
CNQX NAc & systemic --- Yates and Bardo (2017); Yates et al. (2015)
NMDA Receptor
NMDA receptor channel blockers Phencyclidine (PCP) ↑ (lateralized reaction time task) Jentsch & Anzivino (2004)
↑ (5CSRTT) Barnes et al. (2018)
---- (5CSRTT)b Benn & Robinson (2014)
MK-801 (dizocilipne) ↑ (5CSRTT, go/no-go, & rat gambling task) Benn & Robinson (2014); Higgins et al. (2016); Higgins et al. (2018) Isherwood et al. (2015); Onofrychuk et al. (2021); Popik et al. (2015); Terry Jr. et al. (2012)
NS (5CSRTT) Higgins et al. (2005)
--- Higgins et al. (2016); Yates et al. (2015); Yates et al. (2019)
NS Yates et al. (2017a)_
Higgins et al. (2018)
Ketamine ↓ (1CSRTT)c Davis-Reyes et al. (2021)
NS (5CSRTT) Benn & Robinson (2014)
Cottone et al. (2013)
NS Yates et al. (2015); Yates et al. (2017a)
Memantine NS (5CSRTT) Benn & Robinson (2014)
Cottone et al. (2013)
Yates et al. (2017a)
NS
NMDA receptor competitive antagonists AP-5 NAc & systemic --- Cottone et al. (2013); Yates and Bardo (2015)
CGS 19755 Cottone et al. (2013); Yates et al. (2017a)
CPP mPFC ↑ (5CSRTT) Carli et al. (2011)
GluN2B-selective antagonists Ifenprodil NAc & systemic --- (5CSRTT) Higgins et al. (2005)
Yates & Bardo (2017)
↓ (when injected into NAc) Yates et al. (2017a)
NS (when injected systemically)
Ro 25-6981 ↑ (5CSRTT) Higgins et al. (2005)
Ro 63-1908 ↑ (2CSRTT, 5CSRTT, go/no-go, & rat gambling task) Burton & Fletcher, 2012; Higgins et al. (2005); Higgins et al. (2016); Higgins et al. (2018); van der Veen et al. (2021)
Higgins et al. (2016); Yates et al. (2018)
CP-101,606 (traxoprodil) ↑ (5CSRTT & go/no-go) Higgins et al. (2005); Higgins et al. (2016)
--- Higgins et al. (2016); Yates et al. (2018)
Restoration of glutamate homeostasis via N-acetylcysteine ↓ (5CSRTT) Fredricksson et al. (2023)

Abbreviations: AP-5 = (2R)-amino-5-phosphonovaleric acid. CGS 19755 = cis-4-phosphonomethyl-2-piperidine carboxylic acid. CNQX = 6-cyano-7-nitroquinoxaline-2,3-dione. CPP = 3-((+/−)-2-carboxypiperazin-4-yl)propyl-1-phosphonic acid. CSRTT = choice serial reaction time task. DRL = differential reinforcement of low rates of responding. MPEP = 2-Methyl-6-(phenylethynyl)pyridine. MTEP = 3-((2-Methyl-4-thiazolyl)ethynyl)pyridine. NAM = negative allosteric modulator. NS = non-selective drug effects (e.g., increased omissions, response latencies, decreased preference for large magnitude reinforcer when delay is set to 0 s that make interpreting drug-induced changes in impulsive behavior difficult). PAM = positive allosteric modulator.

Notes. The design of the table is based on Table 1 from Pattij and Vanderschuren (2008).

a

In low impulsive rats only.

b

There appears to be some increases in motor impulsivity at 1.0 mg/kg, but this effect disappears at a higher dose (3.0 mg/kg).

c

There was a trend for increased omissions.

Compared to motor impulsivity, the effects of NMDA receptor antagonists on impulsive choice are more nuanced, appearing to be dependent on the specific ligand used and the design of the delay-discounting task. The NMDA receptor competitive antagonist AP-5 fails to alter delay discounting (Cottone et al., 2013; Yates and Bardo, 2017), but the NMDA receptor competitive antagonist CGS 19755 increases impulsive choice (Cottone et al., 2013; Yates et al., 2017a). Similar discrepancies are observed for GluN2B-selective antagonists. Although Ro 63–1908 increases impulsive choice, CP-101,606 does not affect impulsive choice (Yates et al., 2018). Furthermore, ifenprodil leads to decreased preference for a large magnitude reinforcer when its delivery is immediate without selectively altering impulsive choice (Yates et al., 2017a). One of the major challenges of preclinical delay-discounting procedures is dissociating impulsive choice from other constructs such as behavioral flexibility and suboptimal choice (see Yates, 2018 for a full discussion). For example, when the delay to receiving the large, delayed reinforcer systematically increases across the session, the NMDA receptor uncompetitive antagonist MK-801 decreases impulsive choice (Higgins et al., 2018; Yates et al., 2015; Yates et al., 2019; but see Yates et al., 2017a for null effects), which contradicts the effects of NMDA receptor antagonists on motor impulsivity; yet, MK-801 increases impulsive choice when the delay to this alternative decreases across a session (Higgins et al., 2018). This finding indicates that MK-801 increases perseverant responding in this task without necessarily altering impulsive choice. Ketamine and memantine are reported to increase impulsive choice in an adjusting delay procedure (Cottone et al., 2013), but these drugs decrease preference for a large magnitude reinforcer even when the delay to its delivery is set to 0 s (Yates et al., 2015; Yates et al., 2017a), indicating increased suboptimal choice. Thus, the changes in behavior observed following glutamatergic manipulations may not indicate selective alterations in impulsive choice. As such, more work is needed to isolate the influence of glutamate on impulsive choice from behavioral flexibility in preclinical models of delay discounting.

Whereas NMDA receptor antagonists increase motor impulsivity in animals, the NMDA receptor channel blocker memantine improves both hyperactive and inattentive symptoms in adults with ADHD (Surman et al., 2013) and improves total ADHD symptoms as assessed with the ADHD Rating Scale-IV in children, although the effects of memantine on each domain of ADHD are modest (Findling et al., 2007). Ketamine provides rapid symptom improvements for those with MDD (Berman et al., 2000; Fava et al., 2020; Ghasemi et al., 2014; Mandal et al., 2019; Murrough et al., 2013); directly related to the current review, ketamine reduces suicidal ideation (Ballard et al., 2014; Murrough et al., 2015). The discrepancies observed between preclinical and clinical studies most likely stems from the use of outbred rodent strains that do not model aspects of psychiatric conditions. Similar discrepancies are observed for ADHD treatments like d-amphetamine and methylphenidate. These drugs are effective at reducing impulsive behaviors in individuals with ADHD, but they can increase motor impulsivity in healthy individuals (e.g., Voon et al., 2015).

Related to this point, the ability of NMDA receptor antagonists to improve impulsivity in clinical populations may be related to glutamate homeostasis. Glutamate homeostasis is already implicated in substance use disorders (see Kalivas, 2009), and there is increased focus on using N-acetylcysteine, which restores glutamate homeostasis by activating the xC system, for the treatment of various psychiatric disorders (Smaga et al., 2021), although there are few studies testing the effects of N-acetylcysteine on impulsive behaviors. Although not directly tested in individuals with ADHD, N-acetylcysteine improves ADHD Self-Report Scale scores in individuals with lupus (Garcia et al., 2013) and ameliorates motor impulsivity (Fredriksson et al., 2023). The ability of N-acetylcysteine to decrease impulsivity most likely results from increased extrasynaptic glutamate that binds to Group II/III mGluRs on the presynaptic neuron, thus regulating the further release of glutamate into the synapse (see Smaga et al., 2021 for a review). Further supporting this argument is that mGluR2/3 agonists and mGluR2 positive allosteric modulators reduce premature responses in the 5CSRT (van der Veen et al., 2021) and attenuate serotonergic- and glutamatergic-induced increases in motor impulsivity (Pozzi et al., 2011; van der Veen et al., 2021; Wischhof and Koch, 2012; Wischhof et al., 2011) while mGluR2 knockout mice exhibit enhanced locomotor activity (Morishima et al., 2005). The synthetic molecule fasoracetam, which acts as a non-selective mGluR activator, improves ADHD symptom severity in adolescents (Elia et al., 2018). Conversely, mGluR5 antagonists typically fail to alter motor impulsivity and impulsive choice (Isherwood et al., 2015; Semenova and Markou, 2007; Yates et al., 2017b; but see Liu et al., 2008) while mGluR1 antagonists increase motor impulsivity (Sukhotina et al., 2008) and impulsive choice (Yates et al., 2017b; but see Sukhotina et al., 2008).

In contrast to mGluR2 positive allosteric modulators, mGluR4 positive allosteric modulators increase motor impulsivity in mice that exhibit low baseline levels of motor impulsivity in a go/no-go task (Piszczek et al., 2022) and in rats in a 5CSRTT (Isherwood et al., 2017). The discrepancies observed between mGluR2 positive allosteric modulators and mGluR4 positive allosteric modulators may be related to regional differences between these receptor subtypes. mGluR2 mRNA is primarily expressed in the dentate gyrus of the hippocampus and in the cerebellum while mGluR4 expression is highest in cerebellum and in ventrolateral/ventromedial nuclei of the thalamus; mGluR4 mRNA is also higher in parts of the striatum relative to mGluR2 (Testa et al., 1994). Although speculative, these regional differences emphasize the importance of elucidating the neural circuitry involved in glutamatergic control of impulsive behaviors. This is the topic of the next section.

3.5. Glutamatergic Pathways Regulating Impulsive Behaviors

Figure 2 shows glutamatergic projections throughout the brain. There are multiple glutamatergic pathways that originate from the prefrontal cortex (PFC) (see Gasiorowska et al., 2021; Schwartz et al., 2012). The PFC is the most anterior region of the brain and is typically subdivided into lateral PFC (lPFC) and ventromedial PFC (vmPFC), with these regions being further subdivided into dorsolateral PFC (dlPFC), ventrolateral PFC (vlPFC), medial PFC (mPFC), and ventral PFC (vPFC). The orbitofrontal cortex (OFC) is also part of the PFC and is subdivided into medial OFC (mOFC) and lateral OFC (lOFC). Collectively, the structures of the PFC are involved in processes such as language, memory, and executive function (Siddiqui et al., 2008), including decision making (Euston et al., 2012; Hiser and Koenigs, 2018). Not surprisingly, the PFC is a critical mediator of impulsive behaviors, with subregions of the PFC making differential contributions to impulsivity constructs (Kim and Lee, 2010).

Figure 2.

Figure 2.

A schematic depicting glutamatergic projections in the human brain. Abbreviations: ACC = anterior cingulate cortex, Amy = amygdala, DRN = dorsal raphe nuclei, Hip = hippocampus, NAc = nucleus accumbens, PFC = prefrontal cortex, SMA = supplemental motor area, STN = subthalmic nucleus, STR = striatum, TH = thalamus, VTA = ventral tegmental area. Created with BioRender.com.

PFC glutamatergic neurons innervate basal ganglia, specifically dorsal striatum, cerebellum, and motor cortical areas located in the posterior frontal cortex (supplemental motor area, premotor cortex, and primary motor cortex). The basal ganglia are subcortical structures that help facilitate purposeful movement while inhibiting interfering movements (Utter and Basso, 2008) and mediate habit formation (Yin and Knowlton, 2006). Historically believed to regulate fine motor control and balance (Ito, 1984), the cerebellum is now theorized to be involved in multiple higher-order cognitive processes such as executive control and emotional and social processing (Beuriat et al., 2022). Collectively, the motor cortices plan and initiate movements (Nachev et al., 2008; Roland, 1984).

Structures in the limbic system, which are involved in memory, emotion, and motivation, also receive glutamatergic inputs from PFC. The most prominent structures of the limbic system are hippocampus, amygdala, and anterior cingulate cortex (ACC) (Morgane et al., 2005). A critical function of the hippocampus is consolidating short-term memory into long-term memory (Squire et al., 2015). The amygdala is involved in emotional processing (Sergerie et al., 2008), especially fear (Davis, 1992). The ACC is implicated in multiple processes, including conflict monitoring, attention allocation, and decision making (Rolls, 2019). The nucleus accumbens (NAc), a region located in the ventral striatum, is closely related to the limbic system and plays a critical role in reward learning (Day and Carelli, 2007) and the development of addictions (Scofield et al., 2016).

Glutamatergic neurons originating from PFC travel to multiple nuclei located in the brainstem. For the purpose of the current review, I will briefly mention two relevant structures: the substantia nigra and the ventral tegmental area (VTA). The substantia nigra sends dopaminergic neurons to basal ganglia structures, thus serving as a critical mediator of motor control (Luo and Huang, 2016). Stimulation of VTA leads to dopamine release in NAc (Chen, 2022). The final two major glutamate pathways involve the reciprocal connection between PFC and thalamus. The thalamus has numerous functions, such as relaying sensory information to other regions of the brain, regulating sleep, and modulating consciousness (Biesbroek et al., 2024).

Because prefrontal glutamatergic neurons are part of motor control circuits and innervate structures of the limbic system, dysregulation of intra-PFC should be a major contributor of distinct forms of impulsivity. The section below will focus exclusively on aberrations in prefrontal glutamatergic activity that are associated with impulsive behaviors. As impulsivity measures often do not correlate with one another in both humans and animals (e.g., Broos et al., 2012; Marusich et al., 2011; see Strickland and Johnson, 2021 for a review), discussion of how differential dysregulation of glutamatergic pathways accounts for the different types of impulsivity is crucial.

3.5.1. Evidence that Glutamatergic Dysfunction in PFC Mediates Impulsive Behaviors.

Altered prefrontal glutamate levels are often observed in individuals with ADHD, mood disorders, and substance use disorders. Increased glutamate levels in right dlPFC (Bakhshi et al., 2022; MacMaster et al., 2003; but see Carrey et al., 2007), but not in left dlPFC (Bollmann et al., 2015; Puts et al., 2020) are observed in ADHD children, although decreased glutamate levels in right PFC have been reported (Hai et al., 2020a). These inconsistencies may be due to ADHD subtype. For example, children diagnosed with the hyperactive subtype, but not the inattentive subtype, of ADHD have elevated Glx levels in frontal cortex (Courvoisie et al., 2004) while adults primarily diagnosed with the inattentive or the combined subtypes of ADHD have decreased glutamate levels in left midfrontal region (Dramsdahl et al., 2011). When healthy participants are used, there is a positive association between glutamatergic activity in dlPFC and increased motor impulsivity in an auditory go/no-go task (Koizumi et al., 2018). Although individuals with a genetic marker for MDD or bipolar disorder have lower dlPFC glutamate levels (Thomson et al., 2016), increased glutamate/glutamine levels are observed in PFC of individuals diagnosed with bipolar disorder (Castillo et al., 2000; Lan et al., 2009; Michael et al., 2009; but see Smaragdi et al., 2019). Similarly, elevated glutamate levels are observed in the dlPFC of individuals diagnosed with a Cluster B personality disorder, which includes borderline personality disorder and antisocial personality disorder (Smesny et al., 2018). Drug dependence is often associated with elevated glutamate levels in mPFC (Li et al., 2020). Importantly, glutamate levels in mPFC positively correlate with self-reported impulsivity measures in dependent individuals (Li et al., 2020) while glutamate levels in left dlPFC are correlated with cravings for alcohol (Frye et al., 2016). The elevated glutamate observed in mPFC may partially account for why individuals have difficulty inhibiting themselves from using a substance even in the face of negative consequences (e.g., potential for overdose, health complications, legal issues). Indeed, drug craving during abstinence is associated with impulsivity (Doran et al., 2009; Evren et al., 2012; Roozen et al., 2011).

Animal research supports the hypothesis that motor impulsivity is influenced by hyperactive prefrontal glutamatergic activity. Increased excitatory neurotransmitter levels are observed in SHRs in PFC, with evoked glutamate release and glutamate uptake being observed as well (Miller et al., 2014; Rizzo et al., 2017). While SHRs have decreased tonic glutamate signaling in the frontal cortex compared to WKYs, they show increased phasic glutamate signaling (Miller et al., 2019; but see Miller et al., 2014 for a non-statistically significant increase in tonic glutamate signaling in PFC of SHRs), and they show increased glutamate-stimulated release of norepinephrine (Russell, 2001). Intracellular Ca++ levels are elevated in prefrontal cortical neurons of SHRs (Lehohla et al., 2004), which may account for the increased glutamate release observed in these rats. Furthermore, SHRs have increased CAMKII activity in mPFC, which leads to upregulated GluR1 subunit levels (Yabuki et al., 2014). Infusion of NMDA receptor antagonists into infralimbic, but not prelimbic, region of mPFC increases motor impulsivity (Agnoli and Carli, 2012; Benn and Robinson, 2014; Carli et al., 2006; Murphy et al., 2005; Murphy et al., 2012; Pozzi et al., 2011), and preventing the conversion of glutamate to GABA in mPFC increases hyperactivity and increases premature responses in the 5CSRTT (Asinof and Paine, 2013).

Like motor impulsivity, there is evidence that a hyperactive prefrontal glutamatergic system influences affective impulsivity. Increased Glx levels in dlPFC are positively correlated with aggression in individuals with antisocial personality disorder (Smaragdi et al., 2019). Increased Glx levels may be attributed to SNPs in the GAD1 gene that lead to reduced GAD expression, thus leading to accumulating glutamate levels in PFC (Arrúe et al., 2019). Indeed, individuals with bipolar disorder have lower GAD-67 mRNA expression in PFC (Guidotti et al., 2000; Thompson et al., 2009), and they have decreased full-length transcripts, but increased truncated transcripts, of the GAD2 gene in dlPFC (Davis et al., 2016). In addition to altered GAD expression, increased VGLUT-1 and VGLUT-2 expression in dlPFC is observed in women who died by suicide (Powers et al., 2020). Increased VGLUT expression suggests that excess glutamate is being stored in vesicles to be released into the synapse. Men that died by violent suicide have lower EAAT-1 expression than those that died by non-violent suicide, with a similar trend observed for EAAT-2 expression (Powers et al., 2020). These results are consistent with the finding that CSF glutamate levels are positively correlated with suicidal ideations (Garakani et al., 2013). Individuals that have died by suicide, regardless of gender, have increased AMPA receptor (GluR2–4), kainate receptor (subunits 1 and 3), GluN1, GluN2A, GluN2B, mGluR1, mGluR2, and PSD mRNA expression in dlPFC (Gray et al., 2015; Zhao et al., 2018), providing further support for a hyperactive glutamatergic system in individuals prone to engaging in forms of affective impulsivity.

In contrast to motor impulsivity and affective impulsivity, a hypoactive prefrontal glutamatergic system appears to mediate impulsive choice, although the evidence supporting this hypothesis is more indirect compared to other forms of impulsivity. Decreased dlPFC activation is observed in individuals with ADHD during delay-discounting performance (Ortiz et al., 2015), and delay aversion-induced increases in dlPFC activation are observed in male adolescents with ADHD (Van Dessel et al., 2018). The greater activation of dlPFC observed in adolescents with ADHD, relative to controls, may be due to lower baseline levels of excitatory neurotransmission. Children with ADHD, primarily combined subtype, show improvements in impulsive choice following concurrent anodal transcranial direct stimulation of right vmPFC and cathodal stimulation of left dlPFC (Nejati et al., 2021). Specific to bipolar disorder, manic symptoms and depressive symptoms are correlated with BIS-11 attentional impulsivity (Swann et al., 2008). However, mania is positively associated with motor impulsivity while hopelessness is correlated with non-planning impulsivity (Swann et al., 2007; Swann et al., 2008). This result is interesting as non-planning impulsivity is correlated with delay discounting (Baumann and Odum, 2012), and individuals with MDD often show increased impulsive choice in delay-discounting tasks (e.g., Takahashi et al., 2008). Perhaps the lower intra-PFC glutamate levels commonly observed in individuals with MDD (e.g., Ritter et al., 2022; Shirayama et al., 2017) account for increased non-planning impulsivity/impulsive choice.

If hyperactive glutamatergic signaling in PFC contributes to motor/affective impulsivity, the ability of drugs that increase glutamate levels in regions like PFC to improve ADHD symptoms (e.g., memantine; see Section 3.4.) seems paradoxical. One potential explanation to reconcile this discrepancy is that increasing glutamate transmission in PFC restores glutamate homeostasis by allowing glutamate to more easily bind to Group II/Group mGluRs, which regulate the further synthesis and the release of glutamate, thus normalizing glutamate levels in high impulsive individuals. Preclinical research shows that expression of mGluR2 is lower in older adult rats compared to younger adult rats (Hernandez et al., 2018) and decreases in the cortex of mice across development (McOmish et al., 2016). As synaptic pruning of excitatory synapses occurs between adolescence and adulthood (Selemon, 2013), administration of a drug like memantine may allow for glutamate to directly stimulate postsynaptic glutamate receptors, resulting in amelioration of impulsive behaviors in adults with ADHD. This hypothesis is supported by the finding that rats that display increased motor impulsivity have lower mGluR2 mRNA and protein expression and lower mGluR2 receptor binding in mPFC (Elfving et al., 2019; Fomsgaard et al., 2018; Klein et al., 2014). Somewhat relatedly, although systemic administration of lamotrigine decreases reactive aggression (Kumar et al., 2016), lamotrigine fails to ameliorate premature responses following NMDA receptor antagonist infusion into infralimbic cortex (Murphy et al., 2012); in fact, long-term administration of lamotrigine into mPFC increases motor impulsivity (Paine et al., 2015). Long-term lamotrigine administration may lead to a loss of glutamate homeostasis as insufficient glutamate release can prevent the stimulation of extrasynaptic inhibitory mGluRs.

Loss of glutamate homeostasis may also account for altered receptor mRNA/protein expression observed in PFC of high impulsive animals. Despite upregulated GluR1 expression, SHRs have decreased expression of AMPA receptors and reduced AMPA receptor-mediated neurotransmission, with unaltered NMDA receptor-mediated neurotransmission, in PFC (Cheng et al., 2017). Conversely, Roman high avoidance (RHA) rats, a strain of rat selectively bred from Wistar rats (Bignami, 1965) that display enhanced motor impulsivity (Arrondeau et al., 2023; Bellés et al., 2021; Klein et al., 2014), have elevated levels of scaffolding proteins and increased mRNA expression for the GluN2B subunit in PFC compared to Roman low avoidance (RLA) rats (Elfving et al., 2019). Likewise, non-ADHD-like rats demonstrating high motor impulsivity have lower GluN1 and GluN2A, but higher GluN2B, expression in mPFC relative to low impulsive rats (Davis-Reyes et al., 2019). These results are interesting as reduced AMPA receptor-mediated excitatory neurotransmission and increased GluN2B subunit expression indicate weakened synaptic strength (e.g., Kim et al., 2005). At first glance, reduced synaptic strength observed in PFC would suggest a hypoactive glutamatergic system in this region of high impulsive individuals. Indeed, hypoactive PFC is argued to underlie ADHD (see Cheng et al., 2017 for an example). Yet, a hyperactive glutamatergic system in PFC is not necessarily at odds with the hypothesis of hypoactive PFC functioning. Hyperactive glutamatergic signaling can lead to over excitation of inhibitory interneurons that are abundant in the PFC (Ferguson and Gao, 2018), leading to a net result of hypoactive activity. Additionally, excess glutamate causes AMPA receptor internalization (Zhou et al., 2001), which leads to a weakened synapse (Shepherd and Huganir, 2007).

3.5.2. Glutamate and Motor Impulsivity.

Figure 3a shows the glutamatergic pathways implicated in motor impulsivity. The selection and the inhibition of actions are largely controlled by the cortico-basal ganglia-thalamo-cortical loop (Alexander et al., 1986). Movement control is mediated by three distinct pathways in this loop: the direct, the indirect, and the hyperdirect (Rocha et al., 2023). In the direct pathway, dopaminergic neurons originating from the substantia nigra release dopamine in the dorsal striatum, stimulating GABAergic neurons. GABA then binds to GABAergic receptors located in the internal capsule of the globus pallidus. This prevents the globus pallidus from inhibiting the thalamus, allowing the thalamus to innervate the primary motor cortex. Glutamate then activates dopamine D1-like-containing neurons in the dorsal striatum. The indirect pathway also involves dopaminergic transmission from the substantia nigra to the dorsal striatum. However, dopamine binds to inhibitory dopamine D2-like receptors. GABA then binds to GABAergic receptors in the external capsule of the globus pallidus, preventing the inhibition of the subthalamic nucleus. Glutamate binds to NMDA receptors in the internal capsule of the globus pallidus, causing GABA release to inhibit the thalamus. As the thalamus sends glutamatergic neurons to motor cortices (Luo et al., 2019), inhibition of the thalamus leads to insufficient stimulation of the primary motor cortex, preventing movement.

Figure 3.

Figure 3.

Schematics depicting the glutamatergic pathways involved in motor impulsivity (a), impulsive choice (b), and affective impulsivity (c). The red arrows indicate if glutamate levels are increased or decreased in a region. The dashed arrow indicates that the change in glutamate levels is more speculative as additional research is needed. The curved, dashed black arrows indicate that loss of glutamate homeostasis may occur in a particular brain region. In panel a, the purple arrows indicate GABAergic projections. In panel c, the green arrows represent serotonergic projections. Abbreviations: ACC = anterior cingulate cortex, Amy = amygdala, DRN = dorsal raphe nuclei, GP = globus pallidus, Hip = hippocampus, Hyp = hypothalamus, NAc = nucleus accumbens, OFC = orbitofrontal cortex, PFC = prefrontal cortex, SMA = supplemental motor area, STN = subthalmic nucleus, STR = striatum, TH = thalamus, VTA = ventral tegmental area. Note, the changes in glutamate depicted for motor impulsivity (panel a) are similar to what is observed for affective impulsivity. For simplicity, I have focused on changes in corticolimbic regions in panel c. Created with BioRender.com.

Although dopaminergic neurons originating from the substantia nigra are critical mediators of the direct and the indirect pathways, glutamate-stimulated dopamine release is enhanced in the substantia nigra of SHRs (Warton et al., 2009). Furthermore, motor cortical areas project glutamatergic neurons to the dorsal striatum (Paraskevopoulou et al., 2019), and glutamatergic neurons originating from the cerebral cortex modulate activity of the striatum (Gerfen and Surmeier, 2011), including release of dopamine (Cachope and Cheer, 2014); striatal glutamate also mediates the relationship between dopamine synthesis capacity and striatal activation when individuals need to inhibit a prepotent response (Lorenz et al., 2015). Elevated Glx levels in basal ganglia are observed in children with ADHD and bipolar disorder (Bollmann et al., 2015; Carrey et al., 2007; Castillo et al., 2000; MacMaster et al., 2003; but see Naaijen et al., 2016; Puts et al., 2020), and Glx levels decrease in striatum following medication treatment for ADHD (Carrey et al., 2002; Carrey et al., 2003; but see Carrey et al., 2007). Likewise, SHRs have higher striatal glutamine/glutamate levels and have greater evoked glutamate release in striatum (Miller et al., 2014; Rizzo et al., 2017). Additionally, mice lacking actin depolymerizing factor and n-cofilin, which are present in excitatory synapses, have elevated glutamate release in the striatum and display hyperactive/impulsive behaviors that are attenuated by NMDA receptor blockade and methylphenidate administration (Zimmermann et al., 2015).

As in PFC, the increased glutamine levels observed in striatum of ADHD children disappears during adulthood (Bollmann et al., 2015). Adults with ADHD have decreased glutamate levels in basal ganglia, but lower glutamate levels in basal ganglia correlate with more severe symptoms of inattention as opposed to impulsivity (Maltezos et al., 2014). Striatal glutamate levels are lower in animals that display increased motor impulsivity in a DRL task (Chuang et al., 2021). Interestingly, decreased aspartate aminotransferase and increased glutamine synthetase levels are observed in both PFC and striatum of SHRs relative to WKYs, indicating decreased glutamate synthesis and increased glutamate metabolism, respectively (Dimatelis et al., 2015). Similar to what was discussed for PFC, these results implicate an aberrant xC system in striatum, leading to a loss in glutamate homeostasis, which then leads to elevated glutamate levels in striatal regions.

The hyperdirect pathway involves glutamatergic projections from cerebral cortex to subthalamic nucleus. Activity of the internal capsule of the globus pallidus increases, leading to increased inhibition of the thalamus and the primary motor cortex. At first glance, a hyperactive glutamatergic system in the cortico-basal ganglia-thalamo-cortical loop should lead to decreased motor activity, and thus, decreased motor impulsivity. Yet, cerebellar activity also mediates motor control (Jueptner and Weiller, 1998). The deep cerebellar nuclei receive inhibitory inputs from Purkinje cells and excitatory inputs from mossy fiber and climbing fiber pathways (Baumel et al., 2009). The deep cerebellar nuclei innervate the ventrolateral thalamus, which forms a reciprocal loop with motor cortical areas (Ilinky and Kultas-Ilinsky, 2002; Morigaki et al., 2021). While Glx levels in cerebellar cortex are similar in individuals with ADHD and controls (Endres et al., 2015; Rüsch et al., 2010; Soliva et al., 2010; but see Perlov et al., 2010 for increased Glx in left cerebellum of adults with ADHD), children receiving stimulant treatment for ADHD have lower glutamate-glutamine ratios in left cerebellum (Benamor, 2014). This finding is difficult to interpret as most of the participants had the combined subtype (66.67%) or the inattentive subtype (30.39%) of ADHD. Hyperactive cerebellar glutamatergic activity is observed in MDD (Kahl et al., 2020; but see Chen et al., 2014). This hyperactivity may stem from lower GAD-65 and GAD-67 protein levels observed in the cerebellum of individuals with MDD and bipolar disorder (Fatemi et al., 2005; Guidotti et al., 2000). Excess glutamatergic transmission from cerebellum to thalamus would prevent the basal ganglia to adequately inhibit this region, leading to increased movements that interfere with purposeful actions. The result would be increased excitatory outputs from thalamus to primary motor cortex.

There is one potential issue with this interpretation as glutamate/Glx levels are lower in the primary motor cortex of children/adolescents with ADHD (Kahl et al., 2022). One important consideration is that the PFC has reciprocal connections with the premotor cortex (Carmichael and Price, 1995), which itself has reciprocal connections with the primary motor cortex and the supplemental motor area (Ohbayashi, 2021). Hyperpolarization (i.e., reducing activity) of supplemental motor area reduces motor impulsivity in humans (Spieser et al., 2015) whereas impaired inhibitory control is associated with reduced activity of premotor/motor cortex (Weafer et al., 2015). One possible explanation to account for the hypoactive glutamatergic system in premotor/primary cortex is that enhanced glutamatergic transmission from PFC activates inhibitory interneurons in premotor cortex, thus preventing adequate excitation of primary motor cortex. This interpretation is supported by the finding that optogenetic inhibition of premotor inhibitory interneurons increases spontaneous movements in mice (Giordano et al., 2023).

As glutamatergic transmission from primary motor cortex to subthalamic nucleus helps control inhibition of motor actions (Nambu et al., 2002), lower glutamatergic transmission from primary motor cortex may not allow for sufficient stimulation of subthalamic nucleus, resulting in the inability to inhibit competing movements that interfere with the appropriate action (e.g., getting out of a chair during class). This is supported by the finding that use of repetitive transcranial magnetic stimulation to transiently disrupt excitatory neurotransmission in dorsal premotor cortex leads to loss of inhibitory control in healthy participants (Duque et al., 2012). Furthermore, selective reduction of VGLUT in the subthalamic nucleus increases hyperlocomotion, which appears to result from decreased dopamine transporter binding and slower dopamine reuptake (Schweizer et al., 2014). Optogenetic inhibition of subthalamic nucleus increases responses rates during no-go trials in a go/no-go task, indicating increased motor impulsivity; conversely, optogenetic stimulation leads to decreased motor impulsivity (Piszczek et al., 2022).

Beyond the cortico-basal ganglia-thalamo-cortical loop and motor cortices, some limbic structures are implicated in motor impulsivity. The NAc core is connected to the dorsal striatum and controls motor actions related to obtaining reinforcement whereas the shell region mediates the hedonic aspects of reinforcers (Salgado and Kaplitt, 2015). Lesions of the core, but not the shell, increase motor impulsivity (Pothuizen et al., 2005). However, there is evidence that the shell region is involved in motor actions as inactivation of shell selectively increases motor impulsivity (Feja et al., 2014). To further complicate matters, deep brain stimulation of shell increases motor impulsivity whereas stimulation of the core region decreases motor impulsivity (Sesia et al., 2008). These results are difficult to interpret as techniques such as excitotoxic lesions and deep brain stimulation do not isolate the contribution of individual neurotransmitter systems to a behavior. Unfortunately, very few studies have examined the contribution of NAc glutamate to motor impulsivity, but there is evidence that hyperactive glutamatergic activity in NAc mediates motor impulsivity. High impulsive rats, as screened with the 5CSRTT, have reduced GAD-65/67 protein expression; furthermore, selective interference of GAD-65/67 RNA expression in NAc increases motor impulsivity (Caprioli et al., 2014). Tangentially related to this point is that pharmacological disinhibition of NAc via antagonism of GABAergic receptors promotes hyperactivity (Yael et al., 2019).

The hippocampus is another limbic structure implicated in motor impulsivity (Ferland et al., 2014; Mann et al., 2021). There is some evidence that disruption of the glutamatergic system in hippocampus leads to enhanced motor impulsivity. Local genetic deletion of the GluN1 subunit in the CA3 region of the hippocampus increases premature responses in the 5CSRTT while deletion of the GRIA1 gene leads to decreased motor impulsivity (Finlay et al., 2015; Kilonzo et al., 2022). These results indicate that increased AMPA-to-NMDA receptor ratios (i.e., increased LTP resulting from increased excitation) exacerbates motor impulsivity. Increased hippocampal glutamatergic activity could contribute to elevated glutamate levels in NAc as the hippocampus has direct glutamatergic inputs to NAc (e.g., Floresco et al., 2001). The argument for a hyperactive hippocampal glutamatergic system is somewhat challenged by the finding that mice lacking TARP γ-8 exhibit hyperactivity and show reduced AMPA receptor protein expression and AMPA receptor-mediated synaptic transmission in hippocampus (Bai et al., 2022). Downregulation of AMPA receptor protein expression was not observed in PFC of TARP γ-8 knockout mice, but examination of other brain regions was not included. Thus, determining if increased hyperactivity results exclusively from impaired hippocampal AMPA receptor functioning is difficult, especially as motor impulsivity was not tested in conjunction with hyperactivity. As discussed in the next section, a hypoactive hippocampal glutamatergic system is implicated in impulsive choice.

3.5.3. Glutamate and Impulsive Choice.

Impulsive choice is known to be regulated by frontal cortical and limbic regions (Dalley et al., 2011). As discussed in Section 3.5.1., there is evidence that hypoactive intra-PFC glutamatergic activity is associated with impulsive choice. The OFC is another frontal cortical structure that is often studied, but discrepancies are reported in the literature concerning the contribution of OFC to impulsive choice. When whole OFC is lesioned/damaged or pharmacologically inactivated, increases (Berlin et al., 2004; Mobini et al., 2002; Kheramin et al., 2002), decreases (Winstanley et al., 2004), and unaltered (Abela and Chudasama, 2013; Jo et al., 2013; Mariano et al., 2009) impulsive choice are reported. The discrepancies may partially be explained by baseline levels of impulsive choice and the presence of cues during the delay to receiving the large, delayed reinforcer (e.g., Zeeb et al., 2010). More importantly, subregions of OFC have distinct functions, with mOFC representing reward value and lOFC representing punishment and non-reward (see Rolls et al., 2020). However, discrepancies are still observed when a specific region of OFC is targeted. Damage to mOFC increases impulsive choice in humans (Peters and D’Esposito, 2016), but lesions to mOFC decrease impulsive choice in rodents (Mar et al., 2011; but see Stopper et al., 2014 for null effects following pharmacological inactivation). Stronger recruitment of mOFC is associated with increased impulsive choice in individuals with ADHD (Wilbertz et al., 2012), an effect that may be influenced by increased Glx levels in PFC. As the studies cited here did not selectively alter glutamatergic signaling, the contribution of intra-OFC glutamate to impulsive choice needs to be examined. As such, I will limit my discussion to how dysregulation of prefrontal glutamatergic circuits drives impulsive choice (see Fig. 3b).

The PFC exerts top-down control of limbic structures to regulate executive functioning (Del Arco and Mora, 2009). The amygdala has reciprocal connections with both hippocampus (Zhang et al., 2021b) and OFC (Zald and Kim, 1996), and the hippocampus projects neurons back to the OFC (Wikenheiser and Schoenbaum, 2016). As such, corresponding reductions in hippocampal and amygdalar glutamate levels should be associated with impulsive choice. There is evidence to support hypoactive glutamatergic signaling in hippocampus of individuals that display increased impulsive choice. Excitatory neurotransmission is blunted in hippocampus of SHRs (Jensen et al., 2009). Individuals with MDD have decreased hippocampal Glx levels (Block et al., 2009; Zeng et al., 2023; but see Hermens et al., 2015) and decreased VGLUT mRNA expression in hippocampus and surrounding entorhinal cortex and middle temporal gyrus (Medina et al., 2013; Uezato et al., 2009). Even during the euthymic phase (i.e., absence of mood disturbance), increased glutamine and decreased glutamate levels are observed in those with bipolar disorder (Gupta et al., 2022), and decreased VGLUT mRNA expression is observed in entorhinal cortex of individuals with bipolar disorder (Uezato et al., 2009). The lower glutamate levels are not necessarily due to differential EAAT expression as decreased hippocampal expression of EAATs is observed in individuals with MDD, with a similar trend being measured in those with bipolar disorder (Medina et al., 2013). Somewhat similarly, compared to WKYs and Sprague Dawley rats, SHRs have reduced GLT-1 protein expression in hippocampus, but the GLT-1b splice variant is elevated in SHRs in this region (Sterley et al., 2016).

Additional evidence for a hypoactive hippocampal glutamatergic system comes from studies examining ionotropic glutamate receptors. While hypermethylation of the GRIN2A gene is observed in hippocampus of individuals with MDD (Kaut et al., 2015), individuals with bipolar disorder have reduced GluR2, GluR3, and GluR6 mRNA levels in entorhinal cortex (Beneyto et al., 2007), as well as reduced PSD-95 density levels in hippocampus (Toro and Deakin, 2005). SHRs have reduced AMPA receptor-mediated neurotransmission in hippocampus (Medin et al., 2019), but they have increased hippocampal GluN2B expression (Jensen et al., 2009). Furthermore, GluA1 knockout mice show elevated impulsive choice, albeit this effect is modest (Barkus et al., 2012). Although SHRs have higher GluN2B expression, NMDA receptor density is downregulated in both hippocampus and superior temporal cortex in individuals with MDD and bipolar disorder (Nudmamud-Thanoi and Reynolds, 2004; Scarr et al., 2003). Relatedly, reduced hippocampal NMDA receptor functioning is observed in individuals with bipolar disorder (Chitty et al., 2015). Finally, ionotropic glutamate receptor subunit expression (GluR1, GluR3, GluN1, GluN2A, and GluN2B) is downregulated in perirhinal cortex in both MDD and bipolar disorder (Beneyto et al., 2007), a region that has direct connections with the hippocampus (Liu and Bilkey, 1996).

A hypoactive hippocampal glutamatergic system provides an account of the memory disturbances that are commonly observed in individuals with MDD (e.g., Au et al., 2013; Bearden et al., 2006; Cao et al., 2016; Shan et al., 2018; Zaremba et al., 2019) and in SHRs (Grünblatt et al., 2015; Lin et al., 2022). Impaired working memory is linked to impulsive choice (Khurana et al., 2017; Szuhany et al., 2018), and there is some evidence that the hippocampus supports working-memory processes in humans and animals (Borders et al., 2022; Courtney, 2022; Cristoforetti et al., 2022; Hauser et al., 2020; Yoon et al., 2008; but see Slotnick, 2022 for a critical review). Thus, deficient glutamatergic signaling in hippocampus may lead to impaired working memory, which, in turn, can increase impulsive choice in individuals.

While increased decision-making deficits are observed in individuals with amygdalar damage (Gupta et al., 2011), there is not as much evidence supporting hypoactive amygdalar glutamatergic-induced augmentation of impulsive choice. Increased VGLUT protein expression and decreased GAD-67 protein expression are observed in individuals with MDD and bipolar disorder (Varea et al., 2012). These results suggest a hyperactive glutamatergic system, but the increased VGLUT and decreased GAD-67 protein expression may result from deficient glutamate release in the synapse (i.e., compensatory regulation). The reduced glutamate is then unable to effectively regulate amygdalar activity, predisposing at-risk individuals to increased impulsive choice. More recent work using an optogenetic approach to inactivate excitatory neurotransmission during a delay-discounting task reveals complex regulation of impulsive choice by basolateral nucleus of amygdala (BLA). When BLA is inactivated before a choice is made, impulsive choice decreases; however, when this region is inactivated during receipt of a small, immediately delivered reinforcer, impulsive choice increases, but this effect is observed in younger rats only (Hernandez et al., 2019). These results indicate that a hyperactive BLA biases individuals toward immediate reinforcement.

Although there is evidence linking reduced glutamate in hippocampus and, to a lesser extent, amygdala to impulsive choice, enhanced glutamatergic signaling in ACC, which receives inputs from PFC and amygdala (Sarawagi et al., 2021), leads to increased impulsive choice. When glutamate is analyzed in isolation from Glx, adults with ADHD have higher glutamate levels in ACC (Bauer et al., 2018; but see Ende et al., 2016), and women with borderline personality disorder and comorbid ADHD and borderline personality disorder show increased glutamate levels (Hoerst et al., 2010; Rüsch et al., 2010). Glutamate levels are positively correlated with hyperactivity and impulsivity in individuals with ADHD (Bauer et al., 2018). This finding is consistent with the positive correlation between glutamate concentrations in ACC and self-reported impulsive choice (Hoerst et al., 2010), as well as delay discounting in healthy controls (Schmaal et al., 2012a) and in cocaine-dependent individuals (Schmaal et al., 2012b; but see Subramaniam et al., 2022 for null effects with adolescent cannabis users). N-acetylcysteine reduces glutamate levels in the dorsal ACC of cocaine-dependent individuals, with higher levels of BIS-11 impulsivity scores predicting N-acetylcysteine-induced decreases in glutamate (Schmaal et al., 2012b). Presentation of drug-paired stimuli leads to increased intra-ACC glutamate levels (Engeli et al., 2021). These results suggest that increased glutamate in ACC, like PFC, drives impulsive behaviors. The increased ACC glutamatergic activity may occur because hypoactive glutamatergic signaling from PFC is not able to sufficiently stimulate inhibitory parvalbumin interneurons that are highly concentrated in this region (van Heukelum et al., 2019).

Like ACC, increased activity of NAc is associated with impulsive choice in humans (Hamilton et al., 2020; Hariri et al., 2006; Wittmann et al., 2010), and NAc glutamate levels increase following presentation of drug-paired stimuli (Engeli et al., 2021). Paradoxically, SHRs have blunted glutamate-stimulated dopamine release in shell region relative to core region, an effect that is absent in WKYs (Russell, 2003). Inactivating either subregion of NAc increases impulsive choice (Feja et al., 2014), and lesions to the core, but not the shell, increase impulsive choice (Pothuizen et al., 2005). Impulsive choice is also increased in rats with low baseline levels of impulsivity following chemogenetic inhibition of the mPFC-NAc pathway (Wenzel et al., 2023). One potential explanation to reconcile these inconsistent results is that there is a loss of glutamate homeostasis in NAc, which is a central argument of the glutamate homeostasis hypothesis of addiction (Kalivas, 2009). There may be insufficient stimulation of presynaptic mGluRs, resulting in elevated glutamate levels in the NAc that stimulate excitatory receptors (e.g., NDMA). Indeed, Group II and Group III mGluRs are expressed in NAc (Ohishi et al., 1995; Ohishi et al., 1998), and intra-NAc infusion of the GluN2B-selective antagonist ifenprodil, but not the AMPA receptor antagonist CNQX, decreases impulsive choice (Yates and Bardo, 2017). This finding is relevant as GluN2B-containing NDMA receptors are primarily extrasynpatic (Vieira et al., 2020). By blocking GluN2B-containing NMDA receptors, extrasynaptic glutamate can be exchanged for cysteine via the xc system and/or stimulate presynaptic mGluRs.

3.5.4. Glutamate and Affective Impulsivity.

Figure 3c shows the glutamatergic pathways implicated in affective impulsivity. The circuitry involved in affective impulsivity is highly similar to the circuitry controlling motor impulsivity. Hyperactivity in PFC leads to overexcitation of motor control centers, contributing to increased motor impulsivity associated with affective impulsivity (e.g., aggression toward oneself or to another individual). Furthermore, the PFC sends glutamatergic neurons to the dorsal raphe nuclei, where serotonergic cell bodies are located (Michelsen et al., 2008). The raphe nuclei contain GABAergic interneurons (Hernández-Vázquez et al., 2019); as such, increased glutamatergic neurotransmission from PFC leads to increased inhibition of serotoninergic neurons (Soiza-Reilly and Commons, 2011). Serotoninergic hypoactivity is highly associated with suicide (Mann et al., 1990) and aggression (Seo et al., 2008). Deficient serotonin release from the dorsal raphe nuclei leads to disinhibition of the hypothalamus, a region where increased activity is linked to suicide (Bao and Swaab, 2018) and aggression (Hashikawa et al., 2017).

In addition to increased hypothalamic activity, limbic structures become hyperactive. Not only does ACC receive glutamatergic inputs from PFC and amygdala, BLA and mPFC have reciprocal glutamatergic projections (Zimmerman et al., 2019), and the hippocampus sends glutamatergic neurons back to the PFC (Zhou et al., 2020). Descendants of individuals that died by suicide have elevated gene expression for mGluR5 and Homer 1a (García-Gutiérrez et al., 2023). Individuals that died by suicide have increased AMPA receptor (GluR1–4), kainate receptor (subunits 1 and 3), GluN1, GluN2A, GluN2B, mGluR1, mGluR2, VGLUT, and PSD mRNA expression (Zhao et al., 2018), and men that died by suicide have decreased EAAT in ACC (Powers et al., 2020). Relatedly, quinolinic acid levels are increased in ACC of individuals that died by suicide (Steiner et al., 2011). Overexcitation of amygdala, hippocampus, and ACC may lead to excitotoxicity given the large number of ionotropic glutamate receptors in these regions (Monaghan and Cotman, 1985), thus accounting for neuropathology (e.g., atrophy) observed in these regions of individuals that have attempted suicide or in individuals/animals that engage in aggressive behavior (Pardini et al., 2014; Parisi et al., 2021; Roberts et al., 2021; van Heukelum et al., 2021; Zetsche et al., 2007; Zhang et al., 2022; but see Schmaal et al., 2020 for a discussion of inconsistent findings regarding amygdala volume and suicide). Although I did not discuss the ACC as related to motor impulsivity, chemogenetic inhibition of ACC pyramidal cells reduces premature responses in the 5CSRTT (van der Veen et al., 2021). This can be related to affective impulsivity as an individual with a condition like MDD or bipolar disorder may be more likely to engage in self-injurious behavior as a result of a hyperactive ACC.

The increased excitation observed in prefrontal cortical areas and the limbic system accounts for increased NAc activity during aggressive behavior (Chester and DeWall, 2016; Dai et al., 2022; Golden et al., 2019) as glutamatergic inputs mediate dopamine release in this region (Chéramy et al., 1998; Imperato et al., 1990). In addition to receiving glutamatergic inputs from PFC, the NAc receives glutamatergic inputs from hippocampus, amygdala, and ACC (Scofield et al., 2016). Increased NAc activity is also observed when individuals with borderline personality disorder, but not healthy controls, are presented with images of painful stimuli while in a state of heightened distress (Olié et al., 2018). Similarly, individuals that engage in non-suicidal self-injury have increased functional connectivity between NAc and lingual gyrus (in occipital cortex) (Chen et al., 2023), and individuals that have recently attempted suicide have higher NAc volume (Kim et al., 2021). A hyperactive NAc can explain why forms of affective impulsivity, particularly self-harm, are negatively reinforcing (Chapman et al., 2006).

4. Discussion

Glutamate is the most abundant neurotransmitter in the mammalian brain. According to Gasiorowska et al. (2021), over 90% of all neurons express glutamate receptors, and approximately 40% of neurons release glutamate as a neurotransmitter. Considering the ubiquity of glutamate in the central nervous system, there should be little surprise that dysfunctional glutamatergic systems are now implicated in the pathogenesis of numerous conditions like Alzheimer’s disease (Wang and Reddy, 2017), schizophrenia (Kruse and Bustillo, 2022), autism spectrum disorder (Nisar et al., 2022), ADHD (Lesch et al., 2013), mood disorders (Jun et al., 2014), and addiction (Kalivas, 2009; Pettorruso et al., 2014). While the current review focuses on glutamatergic aberrations observed in the last three disorders listed above, impulsive behaviors are commonly observed in each of these conditions, especially impulsive choice (Ahn et al., 2011; Chantiluke et al., 2014; Faja and Dawson, 2015; Geng et al., 2020; Heerey et al., 2007; Thoma et al., 2017; Wang et al., 2020a; Warnell et al., 2019; Yu et al., 2017) and affective impulsivity (Banwari et al., 2013; Caspi, 2015; Fritz et al., 2020; Giannouchos et al., 2023; Kaartinen et al., 2014; Leclerc et al., 2018; Radomsky et al., 1999; Ryden et al., 1991; Serafini et al., 2016; Stark et al., 2022).

4.1. Potential Future Directions

In the current review, I propose that motor impulsivity and affective impulsivity can be attributed to a hyperactive glutamatergic system in PFC that causes downstream dysregulation of motor control regions and limbic system while impulsive choice is characterized by a hypoactive PFC and a combination of hypoactivity and hyperactivity in limbic system. Most of the evidence linking hyperactive glutamatergic systems to impulsive behaviors comes from glutamate/Glx measurements in individuals with a psychiatric disorder and from behavioral pharmacology experiments. One challenge associated with using psychiatric populations is dissociating glutamatergic control of impulsive behavior from other symptoms such as depressed mood. Increased use of nonclinical populations in MRS studies can help minimize this caveat. Yet, one potential limitation associated with MRS studies is differentiating glutamatergic dysfunction from a dysregulated GABA system. The glutamatergic and the GABAergic systems are highly intertwined, with GAD converting glutamate into GABA and GABA transaminase catalyzing GABA into glutamate. Thus, elevated glutamate levels observed in individuals may stem from deficient GABA transmission in interneurons in regions like PFC. Behavioral pharmacology experiments can also be limited as ligands that are commonly used to manipulate the glutamatergic system often have other mechanisms of action. For example, some NMDA receptor antagonists have high affinity for serotonergic and adrenergic receptors (Chenard et al., 1991; Gill et al., 2002; Kapur and Seeman, 2002; Rammes et al., 2001). To circumvent these limitations, additional methods can be used to better elucidate how glutamate drives impulsive behaviors.

Instead of using genetic models of ADHD in which a protein/enzyme has been widely suppressed throughout the central nervous system, a method such as small-interfering RNA (siRNA) can be used to suppress protein expression in a specific region. Alternatively, optogenetic/chemogenetic approaches can be used to selectively infect neurons in brain regions known to have a high expression of glutamatergic neurons. Of the studies cited in the current review, only a few used an approach in which glutamatergic neurons were selectively infected (e.g., van der Veen et al., 2021). Given the concerns about the validity of existing animal models of ADHD such as the SHR (Alsop, 2007; Leffa et al., 2019; Sontag et al., 2010), using genetic approaches can help us further our understanding of the glutamatergic basis of impulsivity.

One emerging field in the treatment of disease is pharmacogenomics (Kabbani et al., 2023). Recent evidence shows that glutamatergic genes are important mediators of treatment outcomes in individuals with conditions like ADHD and bipolar disorder. Specifically, children with the G/G genotype of a specific GRM7 SNP (rs37952452) do not respond as well to methylphenidate treatment relative to children with the G/A genotype (Park et al., 2014). However, individuals with the C/G genotype of the rs2284411 SNP of the GRIN2B gene respond better to methylphenidate treatment (Kim et al., 2017). Using an epigenome pathway analysis, Higgins et al. (2015) show that a glutamatergic network, including CACNG2, CACNA1C, GRIA2, and SLC1A2 genes, mediates lithium response in individuals with bipolar disorder. Increased understanding of the genes involved in impulsive behaviors can help therapists design pharmacological interventions that are highly specific to an individual, which can help improve treatment outcomes.

Considering that glia cells are necessary for glutamatergic functioning, these cells may provide novel approaches for ameliorating impulsive behaviors. As noted in Section 2, glutamine is released from neighboring glia cells, and the xc system is located on astrocytes and microglia (Bridges et al., 2012). Disrupting microglia development during adolescence attenuates high fat diet-induced increases in motor impulsivity later in life (Smith et al., 2020). Exposure to in utero inflammation increases hyperactivity in mice and increased expression of three receptor subtypes that are localized to microglia (Makinson et al., 2019). Mood-stabilizing drugs like lithium cause a downregulation of kainate type 2 receptors located on astrocytes, with no effect on kainate receptors located on neurons (Li et al., 2009), and long-term valproate treatment upregulates EAAT-1 mRNA levels in cultured glia cells (Aguirre et al., 2008). Collectively, these results suggest that decreasing glutamatergic activity at glia cells can improve aspects of impulsivity.

4.2. Conclusions

Both clinical and preclinical studies implicate aberrant glutamatergic systems in the etiology of impulsivity. Specifically, elevated glutamate from prefrontal cortical areas causes hyperactivity of motor control regions, leading to increased motor impulsivity. A hyperactive PFC also leads to over excitation of the limbic system, accounting for forms of affective impulsivity. The shared glutamatergic perturbations observed for motor impulsivity and affective impulsivity can explain why these forms of impulsivity are often correlated with each other (e.g., Armiya’u et al., 2020; Huang et al., 2023; Liu et al., 2019; Park et al., 2020; Pawliczek et al., 2013; Wang et al., 2017a). In contrast to motor/affective impulsivity, a hypoactive PFC is observed in impulsive choice. The differential glutamatergic mechanisms underlying impulsive choice may account for why this form of impulsivity is inconsistently correlated with other measures of impulsivity (e.g., Liu et al., 2019; Stojek et al., 2014; Yan et al., 2016; Yan et al., 2022).

The glutamatergic system is becoming an important target for treating disorders characterized by impulsive behaviors, specifically mood disorders. While seemingly paradoxical, research suggests that an important approach for ameliorating motor impulsivity and/or affective forms of impulsivity in clinical populations is to increase glutamatergic transmission by administering an ionotropic receptor antagonist (e.g., ketamine/memantine). The corresponding increases in glutamate can bind to extrasynaptic Group II/III mGluRs, thus restoring glutamate homeostasis. Alternatively, targeting the cystine-glutamate antiporter directly with N-acetylcysteine may be a promising pharmacotherapy for attenuating impulsive behaviors. Further research targeting the glutamatergic system is important for developing novel interventions for those at risk for engaging in maladaptive impulsive behaviors.

Highlights.

  • Hyperactive glutamate system underlies motor impulsivity and affective impulsivity

  • Hypoactive PFC contributes to impulsive choice

  • Loss of glutamate homeostasis in PFC may lead to motor/affective impulsivity

  • Loss of glutamate homeostasis in nucleus accumbens may mediate impulsive choice

  • Glutamate may be a viable target for treating impulse-control disorders

Acknowledgments

The current work was supported by NIH grant R15DA047610

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Declarations of interest: none

References

  1. Abbott MS, Seaman RW Jr, Doyle MR, Maguire DR, Rice KC, & Collins GT (2022). Interactions between impulsivity and MDPV self-administration in rats. Addiction Biology, 27(3), e13168. 10.1111/adb.13168 [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Abdallah CG, De Feyter HM, Averill LA, Jiang L, Averill CL, Chowdhury GMI, Purohit P, de Graaf RA, Esterlis I, Juchem C, Pittman BP, Krystal JH, Rothman DL, Sanacora G, & Mason GF (2018). The effects of ketamine on prefrontal glutamate neurotransmission in healthy and depressed subjects. Neuropsychopharmacology, 43(10), 2154–2160. 10.1038/s41386-018-0136-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Abe T, Sugihara H, Nawa H, Shigemoto R, Mizuno N, & Nakanishi S (1992). Molecular characterization of a novel metabotropic glutamate receptor mGluR5 coupled to inositol phosphate/Ca2+ signal transduction. The Journal of Biological Chemistry, 267(19), 13361–13368 [PubMed] [Google Scholar]
  4. Abela AR, & Chudasama Y (2013). Dissociable contributions of the ventral hippocampus and orbitofrontal cortex to decision-making with a delayed or uncertain outcome. The European Journal of Neuroscience, 37(4), 640–647. 10.1111/ejn.12071 [DOI] [PubMed] [Google Scholar]
  5. Adams J, Crosbie J, Wigg K, Ickowicz A, Pathare T, Roberts W, Malone M, Schachar R, Tannock R, Kennedy JL, & Barr CL (2004). Glutamate receptor, ionotropic, N-methyl D-aspartate 2A (GRIN2A) gene as a positional candidate for attention-deficit/hyperactivity disorder in the 16p13 region. Molecular Psychiatry, 9(5), 494–499. 10.1038/sj.mp.4001455 [DOI] [PubMed] [Google Scholar]
  6. Adler LA, Kroon RA, Stein M, Shahid M, Tarazi FI, Szegedi A, Schipper J, & Cazorla P (2012). A translational approach to evaluate the efficacy and safety of the novel AMPA receptor positive allosteric modulator org 26576 in adult attention-deficit/hyperactivity disorder. Biological Psychiatry, 72(11), 971–977. 10.1016/j.biopsych.2012.05.012 [DOI] [PubMed] [Google Scholar]
  7. Agnoli L, & Carli M (2012). Dorsal-striatal 5-HT2A and 5-HT2C receptors control impulsivity and perseverative responding in the 5-choice serial reaction time task. Psychopharmacology, 219(2), 633–645. 10.1007/s00213-011-2581-0 [DOI] [PubMed] [Google Scholar]
  8. Agrawal SK, & Fehlings MG (1997). Role of NMDA and non-NMDA ionotropic glutamate receptors in traumatic spinal cord axonal injury. The Journal of Neuroscience, 17(3), 1055–1063. 10.1523/JNEUROSCI.17-03-01055.1997 [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Aguirre G, Rosas S, López-Bayghen E, & Ortega A (2008). Valproate-dependent transcriptional regulation of GLAST/EAAT1 expression: involvement of Ying-Yang 1. Neurochemistry International, 52(7), 1322–1331. 10.1016/j.neuint.2008.01.015 [DOI] [PubMed] [Google Scholar]
  10. Ahn WY, Rass O, Fridberg DJ, Bishara AJ, Forsyth JK, Breier A, Busemeyer JR, Hetrick WP, Bolbecker AR, & O’Donnell BF (2011). Temporal discounting of rewards in patients with bipolar disorder and schizophrenia. Journal of Abnormal Psychology, 120(4), 911–921. 10.1037/a0023333 [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Akutagava-Martins GC, Salatino-Oliveira A, Bruxel EM, Genro JP, Mota NR, Polanczyk GV, Zeni CP, Grevet EH, Bau CH, Rohde LA, & Hutz MH (2014a). Lack of association between the GRM7 gene and attention deficit hyperactivity disorder. Psychiatric Genetics, 24(6), 281–282. 10.1097/YPG.0000000000000059 [DOI] [PubMed] [Google Scholar]
  12. Alexander GE, DeLong MR, & Strick PL (1986). Parallel organization of functionally segregated circuits linking basal ganglia and cortex. Annual Review of Neuroscience, 9, 357–381. 10.1146/annurev.ne.09.030186.002041 [DOI] [PubMed] [Google Scholar]
  13. Aliyev NA, Aliyev ZN, & Aliguliyev AR (1994). Amino acid neurotransmitters in alcohol withdrawal. Alcohol and Alcoholism, 29(6), 643–647. [PubMed] [Google Scholar]
  14. Alsop B (2007). Problems with spontaneously hypertensive rats (SHR) as a model of attention-deficit/hyperactivity disorder (AD/HD). Journal of Neuroscience Methods, 162(1–2), 42–48. 10.1016/j.jneumeth.2006.12.002 [DOI] [PubMed] [Google Scholar]
  15. American Psychiatric Association (2013). Diagnostic and statistical manual of mental disorders (5th ed.). Author. [Google Scholar]
  16. Amitai N, Kuczenski R, Behrens MM, & Markou A (2012). Repeated phencyclidine administration alters glutamate release and decreases GABA markers in the prefrontal cortex of rats. Neuropharmacology, 62(3), 1422–1431. 10.1016/j.neuropharm.2011.01.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Amlung M, Marsden E, Holshausen K, Morris V, Patel H, Vedelago L, Naish KR, Reed DD, & McCabe RE (2019). Delay discounting as a transdiagnostic process in psychiatric disorders: a meta-analysis. JAMA Psychiatry, 76(11), 1176–1186. 10.1001/jamapsychiatry.2019.2102 [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Anwyl R (2009). Metabotropic glutamate receptor-dependent long-term potentiation. Neuropharmacology, 56(4), 735–740. 10.1016/j.neuropharm.2009.01.002 [DOI] [PubMed] [Google Scholar]
  19. Armiya’u AY, Ogunwale A, Bamidele LI, Adole O, & Umar MU (2020). Comparison of impulsivity, aggression and suicidality between prisoners in Nigeria who have committed homicide and those who have not. Criminal Behaviour and Mental Health, 30(5), 240–255. 10.1002/cbm.2161 [DOI] [PubMed] [Google Scholar]
  20. Arrondeau C, Urueña-Méndez G, Bellés L, Marchessaux F, Goutaudier R, & Ginovart N (2023). Motor impulsivity but not risk-related impulsive choice is associated to drug intake and drug-primed relapse. Frontiers in Behavioral Neuroscience, 17, 1200392. 10.3389/fnbeh.2023.1200392 [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Arrúe A, González-Torres MA, Basterreche N, Arnaiz A, Olivas O, Zamalloa MI, Erkoreka L, Catalán A, & Zumárraga M (2019). GAD1 gene polymorphisms are associated with bipolar I disorder and with blood homovanillic acid levels but not with plasma GABA levels. Neurochemistry International, 124, 152–161. 10.1016/j.neuint.2019.01.004 [DOI] [PubMed] [Google Scholar]
  22. Asinof SK, & Paine TA (2013). Inhibition of GABA synthesis in the prefrontal cortex increases locomotor activity but does not affect attention in the 5-choice serial reaction time task. Neuropharmacology, 65, 39–47. 10.1016/j.neuropharm.2012.09.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Au RW, Ungvari GS, Lee E, Man D, Shum DH, Xiang YT, & Tang WK (2013). Prospective memory impairment and its implications for community living skills in bipolar disorder. Bipolar Disorders, 15(8), 885–892. 10.1111/bdi.12122 [DOI] [PubMed] [Google Scholar]
  24. Bai WJ, Luo XG, Jin BH, Zhu KS, Guo WY, Zhu XQ, Qin X, Yang ZX, Zhao JJ, Chen SR, Wang R, Hao J, Wang F, Shi YS, Kong DZ, & Zhang W (2022). Deficiency of transmembrane AMPA receptor regulatory protein γ-8 leads to attention-deficit hyperactivity disorder-like behavior in mice. Zoological Research, 43(5), 851–870. 10.24272/j.issn.2095-8137.2022.122 [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Bakhshi S, Tehrani-Doost M, & Batouli SAH (2022). Evaluation of fronto-cerebellar neurometabolites in youth with ADHD compared to the healthy group and their associations with cognitive and behavioral characteristics: a proton magnetic spectroscopy study. International Journal of Psychophysiology, 182, 190–199. 10.1016/j.ijpsycho.2022.10.005 [DOI] [PubMed] [Google Scholar]
  26. Ballard ED, Ionescu DF, Vande Voort JL, Niciu MJ, Richards EM, Luckenbaugh DA, Brutsché NE, Ameli R, Furey ML, & Zarate CA Jr (2014). Improvement in suicidal ideation after ketamine infusion: relationship to reductions in depression and anxiety. Journal of Psychiatric Research, 58, 161–166. 10.1016/j.jpsychires.2014.07.027 [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Banwari GH, Vankar GK, & Parikh MN (2013). Comparison of suicide attempts in schizophrenia and major depressive disorder: an exploratory study. Asia-Pacific Psychiatry, 5(4), 309–315. 10.1111/j.1758-5872.2012.00188.x [DOI] [PubMed] [Google Scholar]
  28. Bao AM, & Swaab DF (2018). The human hypothalamus in mood disorders: the HPA axis in the center. IBRO Reports, 6, 45–53. 10.1016/j.ibror.2018.11.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Barkus C, Feyder M, Graybeal C, Wright T, Wiedholz L, Izquierdo A, Kiselycznyk C, Schmitt W, Sanderson DJ, Rawlins JN, Saksida LM, Bussey TJ, Sprengel R, Bannerman D, & Holmes A (2012). Do GluA1 knockout mice exhibit behavioral abnormalities relevant to the negative or cognitive symptoms of schizophrenia and schizoaffective disorder?. Neuropharmacology, 62(3), 1263–1272. 10.1016/j.neuropharm.2011.06.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Barnes SA, Young JW, Markou A, Adham N, Gyertyán I, & Kiss B (2018). The effects of cariprazine and aripiprazole on PCP-induced deficits on attention assessed in the 5-choice serial reaction time task. Psychopharmacology, 235(5), 1403–1414. 10.1007/s00213-018-4857-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Barzman DH, DelBello MP, Adler CM, Stanford KE, & Strakowski SM (2006). The efficacy and tolerability of quetiapine versus divalproex for the treatment of impulsivity and reactive aggression in adolescents with co-occurring bipolar disorder and disruptive behavior disorder(s). Journal of Child and Adolescent Psychopharmacology, 16(6), 665–670. 10.1089/cap.2006.16.665 [DOI] [PubMed] [Google Scholar]
  32. Barzman DH, McConville BJ, Masterson B, McElroy S, Sethuraman G, Moore K, Kahwaty AM, & Nelson D (2005). Impulsive aggression with irritability and responsive to divalproex: a pediatric bipolar spectrum disorder phenotype?. Journal of Affective Disorders, 88(3), 279–285. 10.1016/j.jad.2005.08.006 [DOI] [PubMed] [Google Scholar]
  33. Bauer LO, & Covault JM (2020). GRM8 genotype is associated with externalizing disorders and greater inter-trial variability in brain activation during a response inhibition task. Clinical Neurophysiology, 131(6), 1180–1186. 10.1016/j.clinph.2020.02.031 [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Bauer J, Werner A, Kohl W, Kugel H, Shushakova A, Pedersen A, & Ohrmann P (2018). Hyperactivity and impulsivity in adult attention-deficit/hyperactivity disorder is related to glutamatergic dysfunction in the anterior cingulate cortex. The World Journal of Biological Psychiatry, 19(7), 538–546. 10.1080/15622975.2016.1262060 [DOI] [PubMed] [Google Scholar]
  35. Baumann AA, & Odum AL (2012). Impulsivity, risk taking, and timing. Behavioural Processes, 90(3), 408–414. 10.1016/j.beproc.2012.04.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Baumel Y, Jacobson GA, & Cohen D (2009). Implications of functional anatomy on information processing in the deep cerebellar nuclei. Frontiers in Cellular Neuroscience, 3, 14. 10.3389/neuro.03.014.2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Bearden CE, Glahn DC, Monkul ES, Barrett J, Najt P, Villarreal V, & Soares JC (2006). Patterns of memory impairment in bipolar disorder and unipolar major depression. Psychiatry Research, 142(2–3), 139–150. 10.1016/j.psychres.2005.08.010 [DOI] [PubMed] [Google Scholar]
  38. Bellés L, Dimiziani A, Tsartsalis S, Millet P, Herrmann FR, & Ginovart N (2021). Dopamine D2/3 receptor availabilities and evoked dopamine release in striatum differentially predict impulsivity and novelty preference in Roman high- and low-avoidance rats. The International Journal of Neuropsychopharmacology, 24(3), 239–251. 10.1093/ijnp/pyaa084 [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Benamor L (2014). (1)H-Magnetic resonance spectroscopy study of stimulant medication effect on brain metabolites in French Canadian children with attention deficit hyperactivity disorder. Neuropsychiatric Disease and Treatment, 10, 47–54. 10.2147/NDT.S52338 [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Beneyto M, Kristiansen LV, Oni-Orisan A, McCullumsmith RE, & Meador-Woodruff JH (2007). Abnormal glutamate receptor expression in the medial temporal lobe in schizophrenia and mood disorders. Neuropsychopharmacology, 32(9), 1888–1902. 10.1038/sj.npp.1301312 [DOI] [PubMed] [Google Scholar]
  41. Benn A, & Robinson ES (2014). Investigating glutamatergic mechanism in attention and impulse control using rats in a modified 5-choice serial reaction time task. PloS One, 9(12), e115374. 10.1371/journal.pone.0115374 [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Berk M, Plein H, & Ferreira D (2001). Platelet glutamate receptor supersensitivity in major depressive disorder. Clinical Neuropharmacology, 24(3), 129–132. 10.1097/00002826-200105000-00002 [DOI] [PubMed] [Google Scholar]
  43. Berlin HA, Rolls ET, & Kischka U (2004). Impulsivity, time perception, emotion and reinforcement sensitivity in patients with orbitofrontal cortex lesions. Brain, 127(Pt 5), 1108–1126. 10.1093/brain/awh135 [DOI] [PubMed] [Google Scholar]
  44. Berman RM, Cappiello A, Anand A, Oren DA, Heninger GR, Charney DS, & Krystal JH (2000). Antidepressant effects of ketamine in depressed patients. Biological Psychiatry, 47(4), 351–354. 10.1016/s0006-3223(99)00230-9 [DOI] [PubMed] [Google Scholar]
  45. Bettler B, & Mulle C (1995). Review: neurotransmitter receptors. II. AMPA and kainate receptors. Neuropharmacology, 34(2), 123–139. 10.1016/0028-3908(94)00141-e [DOI] [PubMed] [Google Scholar]
  46. Beuriat PA, Cristofori I, Gordon B, & Grafman J (2022). The shifting role of the cerebellum in executive, emotional and social processing across the lifespan. Behavioral and Brain Functions, 18(1), 6. 10.1186/s12993-022-00193-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Biesbroek JM, Verhagen MG, van der Stigchel S, & Biessels GJ (2024). When the central integrator disintegrates: a review of the role of the thalamus in cognition and dementia. Alzheimer’s & Dementia, 20(3), 2209–2222. 10.1002/alz.13563 [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Bignami G (1965). Selection for high rates and low rates of avoidance conditioning in the rat. Animal Behaviour, 13(2), 221–227. 10.1016/0003-3472(65)90038-2 [DOI] [PubMed] [Google Scholar]
  49. Blackstone CD, Moss SJ, Martin LJ, Levey AI, Price DL, & Huganir RL (1992). Biochemical characterization and localization of a non-N-methyl-D-aspartate glutamate receptor in rat brain. Journal of Neurochemistry, 58(3), 1118–1126. 10.1111/j.1471-4159.1992.tb09370.x [DOI] [PubMed] [Google Scholar]
  50. Blader JC, Schooler NR, Jensen PS, Pliszka SR, & Kafantaris V (2009). Adjunctive divalproex versus placebo for children with ADHD and aggression refractory to stimulant monotherapy. The American Journal of Psychiatry, 166(12), 1392–1401. 10.1176/appi.ajp.2009.09020233 [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Bliss TV, & Collingridge GL (1993). A synaptic model of memory: long-term potentiation in the hippocampus. Nature, 361(6407), 31–39. 10.1038/361031a0 [DOI] [PubMed] [Google Scholar]
  52. Block W, Träber F, von Widdern O, Metten M, Schild H, Maier W, Zobel A, & Jessen F (2009). Proton MR spectroscopy of the hippocampus at 3 T in patients with unipolar major depressive disorder: correlates and predictors of treatment response. The International Journal of Neuropsychopharmacology, 12(3), 415–422. 10.1017/S1461145708009516 [DOI] [PubMed] [Google Scholar]
  53. Bollmann S, Ghisleni C, Poil SS, Martin E, Ball J, Eich-Höchli D, Edden RA, Klaver P, Michels L, Brandeis D, & O’Gorman RL (2015). Developmental changes in gamma-aminobutyric acid levels in attention-deficit/hyperactivity disorder. Translational Psychiatry, 5(6), e589. 10.1038/tp.2015.79 [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Borders AA, Ranganath C, & Yonelinas AP (2022). The hippocampus supports high-precision binding in visual working memory. Hippocampus, 32(3), 217–230. 10.1002/hipo.23401 [DOI] [PubMed] [Google Scholar]
  55. Bridges RJ, Natale NR, & Patel SA (2012). System xc cystine/glutamate antiporter: an update on molecular pharmacology and roles within the CNS. British Journal of Pharmacology, 165(1), 20–34. 10.1111/j.1476-5381.2011.01480.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Brogi S, Campiani G, Brindisi M, & Butini S (2019). Allosteric modulation of ionotropic glutamate receptors: an outlook on new therapeutic approaches to treat central nervous system disorders. ACS Medicinal Chemistry Letters, 10(3), 228–236. 10.1021/acsmedchemlett.8b00450 [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Broos N, Schmaal L, Wiskerke J, Kostelijk L, Lam T, Stoop N, Weierink L, Ham J, de Geus EJ, Schoffelmeer AN, van den Brink W, Veltman DJ, de Vries TJ, Pattij T, & Goudriaan AE (2012). The relationship between impulsive choice and impulsive action: a cross-species translational study. PloS One, 7(5), e36781. 10.1371/journal.pone.0036781 [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Bruxel EM, Akutagava-Martins GC, Salatino-Oliveira A, Genro JP, Zeni CP, Polanczyk GV, Chazan R, Schmitz M, Rohde LA, & Hutz MH (2016). GAD1 gene polymorphisms are associated with hyperactivity in Attention-Deficit/Hyperactivity Disorder. American journal of Medical genetics. Part B, Neuropsychiatric Genetics, 171(8), 1099–1104. 10.1002/ajmg.b.32489 [DOI] [PubMed] [Google Scholar]
  59. Burton CL, & Fletcher PJ (2012). Age and sex differences in impulsive action in rats: the role of dopamine and glutamate. Behavioural Brain Research, 230(1), 21–33. 10.1016/j.bbr.2012.01.046 [DOI] [PubMed] [Google Scholar]
  60. Cachope R, & Cheer JF (2014). Local control of striatal dopamine release. Frontiers in Behavioral Neuroscience, 8, 188. 10.3389/fnbeh.2014.00188 [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Cailhol S, & Mormède P (1999). Strain and sex differences in the locomotor response and behavioral sensitization to cocaine in hyperactive rats. Brain Research, 842(1), 200–205. 10.1016/s0006-8993(99)01742-4 [DOI] [PubMed] [Google Scholar]
  62. Cao B, Passos IC, Mwangi B, Bauer IE, Zunta-Soares GB, Kapczinski F, & Soares JC (2016). Hippocampal volume and verbal memory performance in late-stage bipolar disorder. Journal of Psychiatric Research, 73, 102–107. 10.1016/j.jpsychires.2015.12.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Caprioli D, Sawiak SJ, Merlo E, Theobald DE, Spoelder M, Jupp B, Voon V, Carpenter TA, Everitt BJ, Robbins TW, & Dalley JW (2014). Gamma aminobutyric acidergic and neuronal structural markers in the nucleus accumbens core underlie trait-like impulsive behavior. Biological Psychiatry, 75(2), 115–123. 10.1016/j.biopsych.2013.07.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Cardinal RN (2006). Neural systems implicated in delayed and probabilistic reinforcement. Neural Networks, 19(8), 1277–1301. 10.1016/j.neunet.2006.03.004 [DOI] [PubMed] [Google Scholar]
  65. Carli M, Baviera M, Invernizzi RW, & Balducci C (2006). Dissociable contribution of 5-HT1A and 5-HT2A receptors in the medial prefrontal cortex to different aspects of executive control such as impulsivity and compulsive perseveration in rats. Neuropsychopharmacology, 31(4), 757–767. 10.1038/sj.npp.1300893 [DOI] [PubMed] [Google Scholar]
  66. Carli M, Calcagno E, Mainolfi P, Mainini E, & Invernizzi RW (2011). Effects of aripiprazole, olanzapine, and haloperidol in a model of cognitive deficit of schizophrenia in rats: relationship with glutamate release in the medial prefrontal cortex. Psychopharmacology, 214(3), 639–652. 10.1007/s00213-010-2065-7 [DOI] [PubMed] [Google Scholar]
  67. Carli M, Robbins TW, Evenden JL, & Everitt BJ (1983). Effects of lesions to ascending noradrenergic neurones on performance of a 5-choice serial reaction task in rats; implications for theories of dorsal noradrenergic bundle function based on selective attention and arousal. Behavioural Brain Research, 9(3), 361–380. 10.1016/0166-4328(83)90138-9 [DOI] [PubMed] [Google Scholar]
  68. Carmichael ST, & Price JL (1995). Sensory and premotor connections of the orbital and medial prefrontal cortex of macaque monkeys. The Journal of Comparative Neurology, 363(4), 642–664. 10.1002/cne.903630409 [DOI] [PubMed] [Google Scholar]
  69. Carrey N, MacMaster FP, Fogel J, Sparkes S, Waschbusch D, Sullivan S, & Schmidt M (2003). Metabolite changes resulting from treatment in children with ADHD: a 1H-MRS study. Clinical Neuropharmacology, 26(4), 218–221. 10.1097/00002826-200307000-00013 [DOI] [PubMed] [Google Scholar]
  70. Carrey NJ, MacMaster FP, Gaudet L, & Schmidt MH (2007). Striatal creatine and glutamate/glutamine in attention-deficit/hyperactivity disorder. Journal of Child and Adolescent Psychopharmacology, 17(1), 11–17. 10.1089/cap.2006.0008 [DOI] [PubMed] [Google Scholar]
  71. Carrey N, MacMaster FP, Sparkes SJ, Khan SC, & Kusumakar V (2002). Glutamatergic changes with treatment in attention deficit hyperactivity disorder: a preliminary case series. Journal of Child and Adolescent Psychopharmacology, 12(4), 331–336. 10.1089/104454602762599871 [DOI] [PubMed] [Google Scholar]
  72. Carro-Ciampi G, & Bignami G (1968). Effects of scopolamine on shuttle-box avoidance and go-no go discrimination: response-stimulus relationships, pretreatment baselines, and repeated exposure to drug. Psychopharmacologia, 13(2), 89–105. 10.1007/BF00404807 [DOI] [PubMed] [Google Scholar]
  73. Caspi E (2015). Aggressive behaviors between residents with dementia in an assisted living residence. Dementia, 14(4), 528–546. 10.1177/1471301213502588 [DOI] [PubMed] [Google Scholar]
  74. Castillo M, Kwock L, Courvoisie H, & Hooper SR (2000). Proton MR spectroscopy in children with bipolar affective disorder: preliminary observations. AJNR. American Journal of Neuroradiology, 21(5), 832–838. [PMC free article] [PubMed] [Google Scholar]
  75. Catania MV, De Socarraz H, Penney JB, & Young AB (1994). Metabotropic glutamate receptor heterogeneity in rat brain. Molecular Pharmacology, 45(4), 626–636. [PubMed] [Google Scholar]
  76. Chang K, Saxena K, & Howe M (2006). An open-label study of lamotrigine adjunct or monotherapy for the treatment of adolescents with bipolar depression. Journal of the American Academy of Child and Adolescent Psychiatry, 45(3), 298–304. 10.1097/01.chi.0000194566.86160.a3 [DOI] [PubMed] [Google Scholar]
  77. Chantiluke K, Christakou A, Murphy CM, Giampietro V, Daly EM, Ecker C, Brammer M, Murphy DG, MRC AIMS Consortium, & Rubia K (2014). Disorder-specific functional abnormalities during temporal discounting in youth with Attention Deficit Hyperactivity Disorder (ADHD), Autism and comorbid ADHD and Autism. Psychiatry Research, 223(2), 113–120. 10.1016/j.pscychresns.2014.04.006 [DOI] [PubMed] [Google Scholar]
  78. Chapman AL, Gratz KL, & Brown MZ (2006). Solving the puzzle of deliberate self-harm: the experiential avoidance model. Behaviour Research and Therapy, 44(3), 371–394. 10.1016/j.brat.2005.03.005 [DOI] [PubMed] [Google Scholar]
  79. Chatterjee M, Saha S, Dutta N, Sinha S, & Mukhopadhyay K (2022). Kainate receptor subunit 1 (GRIK1) risk variants and GRIK1 deficiency were detected in the Indian ADHD probands. Scientific Reports, 12(1), 18449. 10.1038/s41598-022-21948-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Chen W (2022). Neural circuits provide insights into reward and aversion. Frontiers in Neural Circuits, 16, 1002485. 10.3389/fncir.2022.1002485 [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Chen X, Chen H, Liu J, Tang H, Zhou J, Liu P, Tian Y, Wang X, Lu F, & Zhou J (2023). Functional connectivity alterations in reward-related circuits associated with non-suicidal self-injury behaviors in drug-naïve adolescents with depression. Journal of Psychiatric Research, 163, 270–277. 10.1016/j.jpsychires.2023.05.068 [DOI] [PubMed] [Google Scholar]
  82. Chen LP, Dai HY, Dai ZZ, Xu CT, & Wu RH (2014). Anterior cingulate cortex and cerebellar hemisphere neurometabolite changes in depression treatment: A 1H magnetic resonance spectroscopy study. Psychiatry and Clinical Neurosciences, 68(5), 357–364. 10.1111/pcn.12138 [DOI] [PubMed] [Google Scholar]
  83. Chenard BL, Shalaby IA, Koe BK, Ronau RT, Butler TW, Prochniak MA, Schmidt AW, & Fox CB (1991). Separation of alpha 1 adrenergic and N-methyl-D-aspartate antagonist activity in a series of ifenprodil compounds. Journal of Medicinal Chemistry, 34(10), 3085–3090. 10.1021/jm00114a018 [DOI] [PubMed] [Google Scholar]
  84. Cheng J, Liu A, Shi MY, & Yan Z (2017). Disrupted glutamatergic transmission in prefrontal cortex contributes to behavioral abnormality in an animal model of ADHD. Neuropsychopharmacology, 42(10), 2096–2104. 10.1038/npp.2017.30 [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Chéramy A, L’hirondel M, Godeheu G, Artaud F, & Glowinski J (1998). Direct and indirect presynaptic control of dopamine release by excitatory amino acids. Amino Acids, 14(1–3), 63–68. 10.1007/BF01345244 [DOI] [PubMed] [Google Scholar]
  86. Chester DS, & DeWall CN (2016). The pleasure of revenge: retaliatory aggression arises from a neural imbalance toward reward. Social Cognitive and Affective Neuroscience, 11 7, 1173–82. 10.1093/scan/nsv082 [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Chitty KM, Lagopoulos J, Hickie IB, & Hermens DF (2015). Hippocampal glutamatergic/NMDA receptor functioning in bipolar disorder: a study combining mismatch negativity and proton magnetic resonance spectroscopy. Psychiatry Research, 233(2), 88–94. 10.1016/j.pscychresns.2015.05.002 [DOI] [PubMed] [Google Scholar]
  88. Chuang CY, Tsai SY, Chen SF, Yang YH, Chao CC, Yen NS, & Liao RM (2021). Neurobiological changes in striatal glutamate are associated with trait impulsivity of differential reinforcement of low-rate-response behavior in male rats. Neurobiology of Learning and Memory, 177, 107352. 10.1016/j.nlm.2020.107352 [DOI] [PubMed] [Google Scholar]
  89. Coccaro EF, Lee R, & Vezina P (2013). Cerebrospinal fluid glutamate concentration correlates with impulsive aggression in human subjects. Journal of Psychiatric Research, 47(9), 1247–1253. 10.1016/j.jpsychires.2013.05.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Cole SO, & Michaleski A (1986). Dose-dependent impairment in the performance of a go-no go successive discrimination by chlordiazepoxide. Psychopharmacology, 88(2), 184–186. 10.1007/BF00652237 [DOI] [PubMed] [Google Scholar]
  91. Conn PJ, Christopoulos A, & Lindsley CW (2009). Allosteric modulators of GPCRs: a novel approach for the treatment of CNS disorders. Nature Reviews. Drug Discovery, 8(1), 41–54. 10.1038/nrd2760 [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Cottone P, Iemolo A, Narayan AR, Kwak J, Momaney D, & Sabino V (2013). The uncompetitive NMDA receptor antagonists ketamine and memantine preferentially increase the choice for a small, immediate reward in low-impulsive rats. Psychopharmacology, 226(1), 127–138. 10.1007/s00213-012-2898-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Courtney SM (2022). ‘Working memory is a distributed dynamic process’. Cognitive Neuroscience, 13(3–4), 208–209. 10.1080/17588928.2022.2131747 [DOI] [PubMed] [Google Scholar]
  94. Courvoisie H, Hooper SR, Fine C, Kwock L, & Castillo M (2004). Neurometabolic functioning and neuropsychological correlates in children with ADHD-H: preliminary findings. The Journal of Neuropsychiatry and Clinical Neurosciences, 16(1), 63–69. 10.1176/jnp.16.1.63 [DOI] [PubMed] [Google Scholar]
  95. Cristoforetti G, Majerus S, Sahan MI, van Dijck JP, & Fias W (2022). Neural patterns in parietal cortex and hippocampus distinguish retrieval of start versus end positions in working memory. Journal of Cognitive Neuroscience, 34(7), 1230–1245. 10.1162/jocn_a_01860 [DOI] [PubMed] [Google Scholar]
  96. Cyders MA, & Smith GT (2008). Emotion-based dispositions to rash action: positive and negative urgency. Psychological Bulletin, 134(6), 807–828. 10.1037/a0013341 [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Dai B, Sun F, Tong X, Ding Y, Kuang A, Osakada T, Li Y, & Lin D (2022). Responses and functions of dopamine in nucleus accumbens core during social behaviors. Cell Reports, 40(8), 111246. 10.1016/j.celrep.2022.111246 [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Dalley JW, & Ersche KD (2019). Neural circuitry and mechanisms of waiting impulsivity: relevance to addiction. Philosophical Transactions of the Royal Society of London. Series B, Biological Sciences, 374(1766), 20180145. 10.1098/rstb.2018.0145 [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Dalley JW, Everitt BJ, & Robbins TW (2011). Impulsivity, compulsivity, and top-down cognitive control. Neuron, 69(4), 680–694. 10.1016/j.neuron.2011.01.020 [DOI] [PubMed] [Google Scholar]
  100. Davanzo P, Yue K, Thomas MA, Belin T, Mintz J, Venkatraman TN, Santoro E, Barnett S, & McCracken J (2003). Proton magnetic resonance spectroscopy of bipolar disorder versus intermittent explosive disorder in children and adolescents. The American Journal of Psychiatry, 160(8), 1442–1452. 10.1176/appi.ajp.160.8.1442 [DOI] [PubMed] [Google Scholar]
  101. Davis M (1992). The role of the amygdala in fear and anxiety. Annual Review of Neuroscience, 15, 353–375. 10.1146/annurev.ne.15.030192.002033 [DOI] [PubMed] [Google Scholar]
  102. Davis KN, Tao R, Li C, Gao Y, Gondré-Lewis MC, Lipska BK, Shin JH, Xie B, Ye T, Weinberger DR, Kleinman JE, & Hyde TM (2016). GAD2 Alternative transcripts in the human prefrontal cortex, and in schizophrenia and affective disorders. PloS One, 11(2), e0148558. 10.1371/journal.pone.0148558 [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Davis-Reyes BD, Campbell VM, Land MA, Chapman HL, Stafford SJ, & Anastasio NC (2019). Profile of cortical N-methyl-D-aspartate receptor subunit expression associates with inherent motor impulsivity in rats. Biochemical Pharmacology, 168, 204–213. 10.1016/j.bcp.2019.07.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Davis-Reyes BD, Smith AE, Xu J, Cunningham KA, Zhou J, & Anastasio NC (2021). Subanesthetic ketamine with an AMPAkine attenuates motor impulsivity in rats. Behavioural Pharmacology, 32(4), 335–344. 10.1097/FBP.0000000000000623 [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Day JJ, & Carelli RM (2007). The nucleus accumbens and Pavlovian reward learning. The Neuroscientist, 13(2), 148–159. 10.1177/1073858406295854 [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Del Arco A, & Mora F (2009). Neurotransmitters and prefrontal cortex-limbic system interactions: implications for plasticity and psychiatric disorders. Journal of Neural Transmission, 116(8), 941–952. 10.1007/s00702-009-0243-8 [DOI] [PubMed] [Google Scholar]
  107. Dela Peña I, Shen G, & Shi WX (2021). Droxidopa alters dopamine neuron and prefrontal cortex activity and improves attention-deficit/hyperactivity disorder-like behaviors in rats. European Journal of Pharmacology, 892, 173826. 10.1016/j.ejphar.2020.173826 [DOI] [PubMed] [Google Scholar]
  108. Dickerson F, Stallings C, Vaughan C, Origoni A, Khushalani S, & Yolken R (2012). Antibodies to the glutamate receptor in mania. Bipolar Disorders, 14(5), 547–553. 10.1111/j.1399-5618.2012.01028.x [DOI] [PubMed] [Google Scholar]
  109. Dimatelis JJ, Hsieh JH, Sterley TL, Marais L, Womersley JS, Vlok M, & Russell VA (2015). Impaired energy metabolism and disturbed dopamine and glutamate signalling in the striatum and prefrontal cortex of the Spontaneously Hypertensive Rat model of attention-deficit hyperactivity disorder. Journal of Molecular Neuroscience, 56(3), 696–707. 10.1007/s12031-015-0491-z [DOI] [PubMed] [Google Scholar]
  110. Divito CB, & Underhill SM (2014). Excitatory amino acid transporters: roles in glutamatergic neurotransmission. Neurochemistry International, 73, 172–180. 10.1016/j.neuint.2013.12.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Doran N, Cook J, McChargue D, & Spring B (2009). Impulsivity and cigarette craving: differences across subtypes. Psychopharmacology, 207(3), 365–373. 10.1007/s00213-009-1661-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Dorval KM, Wigg KG, Crosbie J, Tannock R, Kennedy JL, Ickowicz A, Pathare T, Malone M, Schachar R, & Barr CL (2007). Association of the glutamate receptor subunit gene GRIN2B with attention-deficit/hyperactivity disorder. Genes, Brain, and Behavior, 6(5), 444–452. 10.1111/j.1601-183X.2006.00273.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Dramsdahl M, Ersland L, Plessen KJ, Haavik J, Hugdahl K, & Specht K (2011). Adults with attention-deficit/hyperactivity disorder - a brain magnetic resonance spectroscopy study. Frontiers in Psychiatry, 2, 65. 10.3389/fpsyt.2011.00065 [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Dunn GA, Nigg JT, & Sullivan EL (2019). Neuroinflammation as a risk factor for attention deficit hyperactivity disorder. Pharmacology, Biochemistry, and Behavior, 182, 22–34. 10.1016/j.pbb.2019.05.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Duque J, Labruna L, Verset S, Olivier E, & Ivry RB (2012). Dissociating the role of prefrontal and premotor cortices in controlling inhibitory mechanisms during motor preparation. The Journal of Neuroscience, 32(3), 806–816. 10.1523/JNEUROSCI.4299-12.2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Eagle DM, & Robbins TW (2003). Inhibitory control in rats performing a stop-signal reaction-time task: effects of lesions of the medial striatum and d-amphetamine. Behavioral Neuroscience, 117(6), 1302–1317. 10.1037/0735-7044.117.6.1302 [DOI] [PubMed] [Google Scholar]
  117. Elia J, Ungal G, Kao C, Ambrosini A, De Jesus-Rosario N, Larsen L, Chiavacci R, Wang T, Kurian C, Titchen K, Sykes B, Hwang S, Kumar B, Potts J, Davis J, Malatack J, Slattery E, Moorthy G, Zuppa A, Weller A, … Hakonarson H (2018). Fasoracetam in adolescents with ADHD and glutamatergic gene network variants disrupting mGluR neurotransmitter signaling. Nature Communications, 9(1), 4. 10.1038/s41467-017-02244-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Elfving B, Müller HK, Oliveras I, Østerbøg TB, Rio-Alamos C, Sanchez-Gonzalez A, Tobeña A, Fernandez-Teruel A, & Aznar S (2019). Differential expression of synaptic markers regulated during neurodevelopment in a rat model of schizophrenia-like behavior. Progress in Neuro-Psychopharmacology & Biological Psychiatry, 95, 109669. 10.1016/j.pnpbp.2019.109669 [DOI] [PubMed] [Google Scholar]
  119. Ende G, Cackowski S, Van Eijk J, Sack M, Demirakca T, Kleindienst N, Bohus M, Sobanski E, Krause-Utz A, & Schmahl C (2016). Impulsivity and aggression in female BPD and ADHD patients: association with ACC glutamate and GABA concentrations. Neuropsychopharmacology, 41(2), 410–418. 10.1038/npp.2015.153 [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Endres D, Perlov E, Maier S, Feige B, Nickel K, Goll P, Bubl E, Lange T, Glauche V, Graf E, Ebert D, Sobanski E, Philipsen A, & Tebartz van Elst L (2015). Normal neurochemistry in the prefrontal and cerebellar brain of adults with attention-deficit hyperactivity disorder. Frontiers in Behavioral Neuroscience, 9, 242. 10.3389/fnbeh.2015.00242 [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Engeli EJE, Zoelch N, Hock A, Nordt C, Hulka LM, Kirschner M, Scheidegger M, Esposito F, Baumgartner MR, Henning A, Seifritz E, Quednow BB, & Herdener M (2021). Impaired glutamate homeostasis in the nucleus accumbens in human cocaine addiction. Molecular Psychiatry, 26(9), 5277–5285. 10.1038/s41380-020-0828-z [DOI] [PubMed] [Google Scholar]
  122. Enoch MA, Rosser AA, Zhou Z, Mash DC, Yuan Q, & Goldman D (2014). Expression of glutamatergic genes in healthy humans across 16 brain regions; altered expression in the hippocampus after chronic exposure to alcohol or cocaine. Genes, Brain, and Behavior, 13(8), 758–768. 10.1111/gbb.12179 [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Erhardt S, Lim CK, Linderholm KR, Janelidze S, Lindqvist D, Samuelsson M, Lundberg K, Postolache TT, Träskman-Bendz L, Guillemin GJ, & Brundin L (2013). Connecting inflammation with glutamate agonism in suicidality. Neuropsychopharmacology, 38(5), 743–752. 10.1038/npp.2012.248 [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Eriksen J, Li F, & Edwards RH (2020). The mechanism and regulation of vesicular glutamate transport: coordination with the synaptic vesicle cycle. Biochimica et Biophysica Acta. Biomembranes, 1862(12), 183259. 10.1016/j.bbamem.2020.183259 [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Euston DR, Gruber AJ, & McNaughton BL (2012). The role of medial prefrontal cortex in memory and decision making. Neuron, 76(6), 1057–1070. 10.1016/j.neuron.2012.12.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Evenden J (1999). Impulsivity: a discussion of clinical and experimental findings. Journal of Psychopharmacology, 13(2), 180–192. 10.1177/026988119901300211 [DOI] [PubMed] [Google Scholar]
  127. Evenden JL, & Ryan CN (1996). The pharmacology of impulsive behaviour in rats: the effects of drugs on response choice with varying delays of reinforcement. Psychopharmacology, 128(2), 161–170. 10.1007/s002130050121 [DOI] [PubMed] [Google Scholar]
  128. Evren C, Durkaya M, Evren B, Dalbudak E, & Cetin R (2012). Relationship of relapse with impulsivity, novelty seeking and craving in male alcohol-dependent inpatients. Drug and Alcohol Review, 31(1), 81–90. 10.1111/j.1465-3362.2011.00303.x [DOI] [PubMed] [Google Scholar]
  129. Faja S, & Dawson G (2015). Reduced delay of gratification and effortful control among young children with autism spectrum disorders. Autism, 19(1), 91–101. 10.1177/1362361313512424 [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Fatemi SH, Stary JM, Earle JA, Araghi-Niknam M, & Eagan E (2005). GABAergic dysfunction in schizophrenia and mood disorders as reflected by decreased levels of glutamic acid decarboxylase 65 and 67 kDa and Reelin proteins in cerebellum. Schizophrenia Research, 72(2–3), 109–122. 10.1016/j.schres.2004.02.017 [DOI] [PubMed] [Google Scholar]
  131. Fava M, Freeman MP, Flynn M, Judge H, Hoeppner BB, Cusin C, Ionescu DF, Mathew SJ, Chang LC, Iosifescu DV, Murrough J, Debattista C, Schatzberg AF, Trivedi MH, Jha MK, Sanacora G, Wilkinson ST, & Papakostas GI (2020). Double-blind, placebo-controlled, dose-ranging trial of intravenous ketamine as adjunctive therapy in treatment-resistant depression (TRD). Molecular Psychiatry, 25(7), 1592–1603. 10.1038/s41380-018-0256-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Feja M, Hayn L, & Koch M (2014). Nucleus accumbens core and shell inactivation differentially affects impulsive behaviours in rats. Progress in Neuro-Psychopharmacology & Biological Psychiatry, 54, 31–42. 10.1016/j.pnpbp.2014.04.012 [DOI] [PubMed] [Google Scholar]
  133. Ferguson BR, & Gao WJ (2018). PV interneurons: critical regulators of E/I balance for prefrontal cortex-dependent behavior and psychiatric disorders. Frontiers in Neural Circuits, 12, 37. 10.3389/fncir.2018.00037 [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Ferland JM, Zeeb FD, Yu K, Kaur S, Taves MD, & Winstanley CA (2014). Greater sensitivity to novelty in rats is associated with increased motor impulsivity following repeated exposure to a stimulating environment: implications for the etiology of impulse control deficits. The European Journal of Neuroscience, 40(12), 3746–3756. 10.1111/ejn.12748 [DOI] [PubMed] [Google Scholar]
  135. Ferster CB, & Skinner BF (1957). Schedules of reinforcement. Appleton-Century-Crofts. 10.1037/10627-000 [DOI] [Google Scholar]
  136. Findling RL, McNamara NK, Stansbrey RJ, Maxhimer R, Periclou A, Mann A, & Graham SM (2007). A pilot evaluation of the safety, tolerability, pharmacokinetics, and effectiveness of memantine in pediatric patients with attention-deficit/hyperactivity disorder combined type. Journal of Child and Adolescent Psychopharmacology, 17(1), 19–33. 10.1089/cap.2006.0044 [DOI] [PubMed] [Google Scholar]
  137. Finlay JM, Dunham GA, Isherwood AM, Newton CJ, Nguyen TV, Reppar PC, Snitkovski I, Paschall SA, & Greene RW (2015). Effects of prefrontal cortex and hippocampal NMDA NR1-subunit deletion on complex cognitive and social behaviors. Brain Research, 1600, 70–83. 10.1016/j.brainres.2014.10.037 [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Floresco SB, Todd CL, & Grace AA (2001). Glutamatergic afferents from the hippocampus to the nucleus accumbens regulate activity of ventral tegmental area dopamine neurons. The Journal of Neuroscience, 21(13), 4915–4922. 10.1523/JNEUROSCI.21-13-04915.2001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  139. Fomsgaard L, Moreno JL, de la Fuente Revenga M, Brudek T, Adamsen D, Rio-Alamos C, Saunders J, Klein AB, Oliveras I, Cañete T, Blazquez G, Tobeña A, Fernandez-Teruel A, Gonzalez-Maeso J, & Aznar S (2018). Differences in 5-HT2A and mGlu2 receptor expression levels and repressive epigenetic modifications at the 5-HT2A promoter region in the Roman low- (RLA-I) and high- (RHA-I) avoidance rat strains. Molecular Neurobiology, 55(3), 1998–2012. 10.1007/s12035-017-0457-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Fox AT, Hand DJ, & Reilly MP (2008). Impulsive choice in a rodent model of attention-deficit/hyperactivity disorder. Behavioural Brain Research, 187(1), 146–152. 10.1016/j.bbr.2007.09.008 [DOI] [PubMed] [Google Scholar]
  141. Fox AE, Nicholson AM, Singha D, Thieret BAS, Ortiz M, & Visser EJ (2023). Timing and delay discounting in attention-deficit/hyperactivity disorder: A translational approach. Developmental Psychobiology, 65(5), e22399. 10.1002/dev.22399 [DOI] [PubMed] [Google Scholar]
  142. Fredriksson A, & Archer T (2003). Hyperactivity following postnatal NMDA antagonist treatment: reversal by D-amphetamine. Neurotoxicity Research, 5(7), 549–564. 10.1007/BF03033165 [DOI] [PubMed] [Google Scholar]
  143. Fredriksson I, Jayaram-Lindström N, Kalivas PW, Melas PA, & Steensland P (2023). N-acetylcysteine improves impulse control and attenuates relapse-like alcohol intake in long-term drinking rats. Behavioural Brain Research, 436, 114089. 10.1016/j.bbr.2022.114089 [DOI] [PubMed] [Google Scholar]
  144. Fritz M, Shenar R, Cardenas-Morales L, Jäger M, Streb J, Dudeck M, & Franke I (2020). Aggressive and disruptive behavior among psychiatric patients with major depressive disorder, schizophrenia, or alcohol dependency and the effect of depression and self-esteem on aggression. Frontiers in Psychiatry, 11, 599828. 10.3389/fpsyt.2020.599828 [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Frye MA, Hinton DJ, Karpyak VM, Biernacka JM, Gunderson LJ, Geske J, Feeder SE, Choi DS, & Port JD (2016). Elevated glutamate levels in the left dorsolateral prefrontal cortex are associated with higher cravings for alcohol. Alcoholism: Clinical and Experimental Research, 40(8), 1609–1616. 10.1111/acer.13131 [DOI] [PubMed] [Google Scholar]
  146. Frye MA, Tsai GE, Huggins T, Coyle JT, & Post RM (2007a). Low cerebrospinal fluid glutamate and glycine in refractory affective disorder. Biological Psychiatry, 61(2), 162–166. 10.1016/j.biopsych.2006.01.024 [DOI] [PubMed] [Google Scholar]
  147. Galea I (2021). The blood-brain barrier in systemic infection and inflammation. Cellular & Molecular Immunology, 18(11), 2489–2501. 10.1038/s41423-021-00757-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  148. Gan Q, Salussolia CL, & Wollmuth LP (2015). Assembly of AMPA receptors: mechanisms and regulation. The Journal of Physiology, 593(1), 39–48. 10.1113/jphysiol.2014.273755 [DOI] [PMC free article] [PubMed] [Google Scholar]
  149. Garakani A, Martinez JM, Yehuda R, & Gorman JM (2013). Cerebrospinal fluid levels of glutamate and corticotropin releasing hormone in major depression before and after treatment. Journal of Affective Disorders, 146(2), 262–265. 10.1016/j.jad.2012.06.037 [DOI] [PubMed] [Google Scholar]
  150. Garcia RJ, Francis L, Dawood M, Lai ZW, Faraone SV, & Perl A (2013). Attention deficit and hyperactivity disorder scores are elevated and respond to N-acetylcysteine treatment in patients with systemic lupus erythematosus. Arthritis and Rheumatism, 65(5), 1313–1318. 10.1002/art.37893 [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. García-Gutiérrez MS, Navarro D, Torregrosa AB, Viudez-Martínez A, Giner S, & Manzanares J (2023). Alterations of BDNF, mGluR5, Homer1a, p11 and excitatory/inhibitory balance in corticolimbic brain regions of suicide decedents. Journal of Affective Disorders, 339, 366–376. 10.1016/j.jad.2023.07.003 [DOI] [PubMed] [Google Scholar]
  152. Gasiorowska A, Wydrych M, Drapich P, Zadrozny M, Steczkowska M, Niewiadomski W, & Niewiadomska G (2021). The biology and pathobiology of glutamatergic, cholinergic, and dopaminergic signaling in the aging brain. Frontiers in Aging Neuroscience, 13, 654931. 10.3389/fnagi.2021.654931 [DOI] [PMC free article] [PubMed] [Google Scholar]
  153. Gasparini F, & Spooren W (2007). Allosteric modulators for mGlu receptors. Current Neuropharmacology, 5(3), 187–194. 10.2174/157015907781695900 [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. Geng Z, Wu X, Wang L, Zhou S, Tian Y, Wang K, & Wei L (2020). Reduced delayed reward selection by Alzheimer’s disease and mild cognitive impairment patients during intertemporal decision-making. Journal of Clinical and Experimental Neuropsychology, 42(3), 298–306. 10.1080/13803395.2020.1711873 [DOI] [PubMed] [Google Scholar]
  155. Gerfen CR, & Surmeier DJ (2011). Modulation of striatal projection systems by dopamine. Annual Review of Neuroscience, 34, 441–466. 10.1146/annurev-neuro-061010-113641 [DOI] [PMC free article] [PubMed] [Google Scholar]
  156. Ghasemi M, Kazemi MH, Yoosefi A, Ghasemi A, Paragomi P, Amini H, & Afzali MH (2014). Rapid antidepressant effects of repeated doses of ketamine compared with electroconvulsive therapy in hospitalized patients with major depressive disorder. Psychiatry Research, 215(2), 355–361. 10.1016/j.psychres.2013.12.008 [DOI] [PubMed] [Google Scholar]
  157. Giannouchos TV, Beverly J, Christodoulou I, & Callaghan T (2023). Suicide and non-fatal self-injury-related emergency department visits among individuals with autism spectrum disorder. Autism, 27(7), 1983–1996. 10.1177/13623613221150089 [DOI] [PubMed] [Google Scholar]
  158. Gill R, Alanine A, Bourson A, Buttelmann B, Fischer G, Heitz MP, Kew JN, Levet-Trafit B, Lorez HP, Malherbe P, Miss MT, Mutel V, Pinard E, Roever S, Schmitt M, Trube G, Wybrecht R, Wyler R, & Kemp JA (2002). Pharmacological characterization of Ro 63–1908 (1-[2-(4-hydroxy-phenoxy)-ethyl]-4-(4-methyl-benzyl)-piperidin-4-ol), a novel subtype-selective N-methyl-D-aspartate antagonist. The Journal of Pharmacology and Experimental Therapeutics, 302(3), 940–948. 10.1124/jpet.102.034322 [DOI] [PubMed] [Google Scholar]
  159. Giordano N, Alia C, Fruzzetti L, Pasquini M, Palla G, Mazzoni A, Micera S, Fogassi L, Bonini L, & Caleo M (2023). Fast-spiking interneurons of the premotor cortex contribute to initiation and execution of spontaneous actions. The Journal of Neuroscience, 43(23), 4234–4250. 10.1523/JNEUROSCI.0750-22.2023 [DOI] [PMC free article] [PubMed] [Google Scholar]
  160. Gladding CM, Fitzjohn SM, & Molnár E (2009). Metabotropic glutamate receptor-mediated long-term depression: molecular mechanisms. Pharmacological Reviews, 61(4), 395–412. 10.1124/pr.109.001735 [DOI] [PMC free article] [PubMed] [Google Scholar]
  161. Glessner JT, Khan ME, Chang X, Liu Y, Otieno FG, Lemma M, Slaby I, Hain H, Mentch F, Li J, Kao C, Sleiman PMA, March ME, Connolly J, & Hakonarson H (2023). Rare recurrent copy number variations in metabotropic glutamate receptor interacting genes in children with neurodevelopmental disorders. Journal of Neurodevelopmental Disorders, 15(1), 14. 10.1186/s11689-023-09483-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  162. Golden SA, Jin M, Heins C, Venniro M, Michaelides M, & Shaham Y (2019). Nucleus accumbens Drd1-expressing neurons control aggression self-administration and aggression seeking in mice. The Journal of Neuroscience, 39(13), 2482–2496. 10.1523/JNEUROSCI.2409-18.2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Gray AL, Hyde TM, Deep-Soboslay A, Kleinman JE, & Sodhi MS (2015). Sex differences in glutamate receptor gene expression in major depression and suicide. Molecular Psychiatry, 20(9), 1057–1068. 10.1038/mp.2015.91 [DOI] [PubMed] [Google Scholar]
  164. Green L, Myerson J, & Calvert AL (2010). Pigeons’ discounting of probabilistic and delayed reinforcers. The Journal of the Experimental Analysis of Behavior, 94(2), 113–123. 10.1901/jeab.2010.94-113 [DOI] [PMC free article] [PubMed] [Google Scholar]
  165. Grünblatt E, Bartl J, Iuhos DI, Knezovic A, Trkulja V, Riederer P, Walitza S, & Salkovic-Petrisic M (2015). Characterization of cognitive deficits in spontaneously hypertensive rats, accompanied by brain insulin receptor dysfunction. Journal of Molecular Psychiatry, 3(1), 6. 10.1186/s40303-015-0012-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  166. Guidotti A, Auta J, Davis JM, Di-Giorgi-Gerevini V, Dwivedi Y, Grayson DR, Impagnatiello F, Pandey G, Pesold C, Sharma R, Uzunov D, & Costa E (2000). Decrease in reelin and glutamic acid decarboxylase67 (GAD67) expression in schizophrenia and bipolar disorder: a postmortem brain study. Archives of General Psychiatry, 57(11), 1061–1069. 10.1001/archpsyc.57.11.1061 [DOI] [PubMed] [Google Scholar]
  167. Gulsun M, Oznur T, Aydemir E, Ozcelik F, Erdem M, Zincir S, Akgul O, & Kurt Y (2016). Possible relationship between amino acids, aggression and psychopathy. International Journal of Psychiatry in Clinical Practice, 20(2), 91–100. 10.3109/13651501.2016.1144771 [DOI] [PubMed] [Google Scholar]
  168. Gungor Aydin A, & Adiguzel E (2023). The mesocortical dopaminergic system cannot explain hyperactivity in an animal model of attention deficit hyperactivity disorder (ADHD)- Spontaneously hypertensive rats (SHR). Laboratory Animal Research, 39(1), 20. 10.1186/s42826-023-00172-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  169. Gupta R, Koscik TR, Bechara A, & Tranel D (2011). The amygdala and decision-making. Neuropsychologia, 49(4), 760–766. 10.1016/j.neuropsychologia.2010.09.029 [DOI] [PMC free article] [PubMed] [Google Scholar]
  170. Gupta R, Sood M, Sharma U, Bhargava R, Jagannathan NR, & Chadda RK (2022). Neurochemical correlates of cognitive functions in euthymic patients with bipolar disorder: 1H-MRS study. Asian Journal of Psychiatry, 78, 103273. 10.1016/j.ajp.2022.103273 [DOI] [PubMed] [Google Scholar]
  171. Gvion Y, Levi-Belz Y, Hadlaczky G, & Apter A (2015). On the role of impulsivity and decision-making in suicidal behavior. World Journal of Psychiatry, 5(3), 255–259. 10.5498/wjp.v5.i3.255 [DOI] [PMC free article] [PubMed] [Google Scholar]
  172. Hai T, Duffy H, Lemay JF, Swansburg R, Climie EA, & MacMaster FP (2020a). Neurochemical correlates of executive function in children with attention-deficit/hyperactivity disorder. Journal of the Canadian Academy of Child and Adolescent Psychiatry, 29(1), 15–25. [PMC free article] [PubMed] [Google Scholar]
  173. Hamilton KR, Smith JF, Gonçalves SF, Nketia JA, Tasheuras ON, Yoon M, Rubia K, Chirles TJ, Lejuez CW, & Shackman AJ (2020). Striatal bases of temporal discounting in early adolescents. Neuropsychologia, 144, 107492. 10.1016/j.neuropsychologia.2020.107492 [DOI] [PMC free article] [PubMed] [Google Scholar]
  174. Hankosky ER, & Gulley JM (2013). Performance on an impulse control task is altered in adult rats exposed to amphetamine during adolescence. Developmental Psychobiology, 55(7), 733–744. 10.1002/dev.21067 [DOI] [PMC free article] [PubMed] [Google Scholar]
  175. Hariri AR, Brown SM, Williamson DE, Flory JD, de Wit H, & Manuck SB (2006). Preference for immediate over delayed rewards is associated with magnitude of ventral striatal activity. The Journal of Neuroscience, 26(51), 13213–13217. 10.1523/JNEUROSCI.3446-06.2006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  176. Hashikawa Y, Hashikawa K, Falkner AL, & Lin D (2017). Ventromedial hypothalamus and the generation of aggression. Frontiers in Systems Neuroscience, 11, 94. 10.3389/fnsys.2017.00094 [DOI] [PMC free article] [PubMed] [Google Scholar]
  177. Hashimoto K, Bruno D, Nierenberg J, Marmar CR, Zetterberg H, Blennow K, & Pomara N (2016). Abnormality in glutamine-glutamate cycle in the cerebrospinal fluid of cognitively intact elderly individuals with major depressive disorder: a 3-year follow-up study. Translational Psychiatry, 6(3), e744. 10.1038/tp.2016.8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  178. Hauser J, Llano López LH, Feldon J, Gargiulo PA, & Yee BK (2020). Small lesions of the dorsal or ventral hippocampus subregions are associated with distinct impairments in working memory and reference memory retrieval, and combining them attenuates the acquisition rate of spatial reference memory. Hippocampus, 30(9), 938–957. 10.1002/hipo.23207 [DOI] [PubMed] [Google Scholar]
  179. Heerey EA, Robinson BM, McMahon RP, & Gold JM (2007). Delay discounting in schizophrenia. Cognitive Neuropsychiatry, 12(3), 213–221. 10.1080/13546800601005900 [DOI] [PMC free article] [PubMed] [Google Scholar]
  180. Hermens DF, Chitty KM, Lee RS, Tickell A, Haber PS, Naismith SL, Hickie IB, & Lagopoulos J (2015). Hippocampal glutamate is increased and associated with risky drinking in young adults with major depression. Journal of Affective Disorders, 186, 95–98. 10.1016/j.jad.2015.07.009 [DOI] [PubMed] [Google Scholar]
  181. Hernandez CM, McQuail JA, Schwabe MR, Burke SN, Setlow B, & Bizon JL (2018). Age-related declines in prefrontal cortical expression of metabotropic glutamate receptors that support working memory. eNeuro, 5(3), ENEURO.0164–18.2018. 10.1523/ENEURO.0164-18.2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  182. Hernandez CM, Orsini CA, Labiste CC, Wheeler AR, Ten Eyck TW, Bruner MM, Sahagian TJ, Harden SW, Frazier CJ, Setlow B, & Bizon JL (2019). Optogenetic dissection of basolateral amygdala contributions to intertemporal choice in young and aged rats. eLife, 8, e46174. 10.7554/eLife.46174 [DOI] [PMC free article] [PubMed] [Google Scholar]
  183. Hernández-Vázquez F, Garduño J, & Hernández-López S (2019). GABAergic modulation of serotonergic neurons in the dorsal raphe nucleus. Reviews in the Neurosciences, 30(3), 289–303. 10.1515/revneuro-2018-0014 [DOI] [PubMed] [Google Scholar]
  184. Herring BE, & Nicoll RA (2016). Long-term potentiation: from CaMKII to AMPA receptor trafficking. Annual Review of Physiology, 78, 351–365. 10.1146/annurev-physiol-021014-071753 [DOI] [PubMed] [Google Scholar]
  185. Higgins GA, Allyn-Feuer A, Barbour E, & Athey BD (2015). A glutamatergic network mediates lithium response in bipolar disorder as defined by epigenome pathway analysis. Pharmacogenomics, 16(14), 1547–1563. 10.2217/pgs.15.106 [DOI] [PubMed] [Google Scholar]
  186. Higgins GA, Ballard TM, Enderlin M, Haman M, & Kemp JA (2005). Evidence for improved performance in cognitive tasks following selective NR2B NMDA receptor antagonist pre-treatment in the rat. Psychopharmacology, 179(1), 85–98. 10.1007/s00213-005-2203-9 [DOI] [PubMed] [Google Scholar]
  187. Higgins GA, Silenieks LB, MacMillan C, Sevo J, Zeeb FD, & Thevarkunnel S (2016). Enhanced attention and impulsive action following NMDA receptor GluN2B-selective antagonist pretreatment. Behavioural Brain Research, 311, 1–14. 10.1016/j.bbr.2016.05.025 [DOI] [PubMed] [Google Scholar]
  188. Higgins GA, Silenieks LB, MacMillan C, Zeeb FD, & Thevarkunnel S (2018). Effects of the NMDA receptor antagonists dizocilpine and Ro 63–1908 on delay-discounting and risky decision-making in a gambling task. Behavioural Brain Research, 348, 201–210. 10.1016/j.bbr.2018.04.028 [DOI] [PubMed] [Google Scholar]
  189. Hiser J, & Koenigs M (2018). The multifaceted role of the ventromedial prefrontal cortex in emotion, decision making, social cognition, and psychopathology. Biological Psychiatry, 83(8), 638–647. 10.1016/j.biopsych.2017.10.030 [DOI] [PMC free article] [PubMed] [Google Scholar]
  190. Hoerst M, Weber-Fahr W, Tunc-Skarka N, Ruf M, Bohus M, Schmahl C, & Ende G (2010). Correlation of glutamate levels in the anterior cingulate cortex with self-reported impulsivity in patients with borderline personality disorder and healthy controls. Archives of General Psychiatry, 67(9), 946–954. 10.1001/archgenpsychiatry.2010.93 [DOI] [PubMed] [Google Scholar]
  191. Hollander E, Swann AC, Coccaro EF, Jiang P, & Smith TB (2005). Impact of trait impulsivity and state aggression on divalproex versus placebo response in borderline personality disorder. The American Journal of Psychiatry, 162(3), 621–624. 10.1176/appi.ajp.162.3.621 [DOI] [PubMed] [Google Scholar]
  192. Holton KF, Johnstone JM, Brandley ET, & Nigg JT (2019). Evaluation of dietary intake in children and college students with and without attention-deficit/hyperactivity disorder. Nutritional Neuroscience, 22(9), 664–677. 10.1080/1028415X.2018.1427661 [DOI] [PMC free article] [PubMed] [Google Scholar]
  193. Hoptman MJ (2015). Impulsivity and aggression in schizophrenia: a neural circuitry perspective with implications for treatment. CNS Spectrums, 20(3), 280–286. 10.1017/S1092852915000206 [DOI] [PMC free article] [PubMed] [Google Scholar]
  194. Huang MH, Kuan YH, Tu PC, Chang WC, Chan YE, & Su TP (2023). Brain structural abnormalities and trait impulsivity in suicidal and non-suicidal patients with bipolar disorder. Journal of Affective Disorders, 333, 10–17. 10.1016/j.jad.2023.04.050 [DOI] [PubMed] [Google Scholar]
  195. Hudson MR, Sokolenko E, O’Brien TJ, & Jones NC (2020). NMDA receptors on parvalbumin-positive interneurons and pyramidal neurons both contribute to MK-801 induced gamma oscillatory disturbances: complex relationships with behaviour. Neurobiology of Disease, 134, 104625. 10.1016/j.nbd.2019.104625 [DOI] [PubMed] [Google Scholar]
  196. Ilinsky IA, & Kultas-Ilinsky K (2002). Motor thalamic circuits in primates with emphasis on the area targeted in treatment of movement disorders. Movement Disorders, 17 Suppl 3, S9–S14. 10.1002/mds.10137 [DOI] [PubMed] [Google Scholar]
  197. Imperato A, Honoré T, & Jensen LH (1990). Dopamine release in the nucleus caudatus and in the nucleus accumbens is under glutamatergic control through non-NMDA receptors: a study in freely-moving rats. Brain Research, 530(2), 223–228. 10.1016/0006-8993(90)91286-p [DOI] [PubMed] [Google Scholar]
  198. Inoshita M, Umehara H, Watanabe SY, Nakataki M, Kinoshita M, Tomioka Y, Tajima A, Numata S, & Ohmori T (2018). Elevated peripheral blood glutamate levels in major depressive disorder. Neuropsychiatric Disease and Treatment, 14, 945–953. 10.2147/NDT.S159855 [DOI] [PMC free article] [PubMed] [Google Scholar]
  199. Irimia C, Buczynski MW, Natividad LA, Laredo SA, Avalos N, & Parsons LH (2017). Dysregulated glycine signaling contributes to increased impulsivity during protracted alcohol abstinence. The Journal of Neuroscience, 37(7), 1853–1861. 10.1523/JNEUROSCI.2466-16.2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  200. Irimia C, Tuong RN, Quach T, & Parsons LH (2014). Impaired response inhibition in the rat 5 choice continuous performance task during protracted abstinence from chronic alcohol consumption. PloS One, 9(10), e109948. 10.1371/journal.pone.0109948 [DOI] [PMC free article] [PubMed] [Google Scholar]
  201. Irimia C, Wiskerke J, Natividad LA, Polis IY, de Vries TJ, Pattij T, & Parsons LH (2015). Increased impulsivity in rats as a result of repeated cycles of alcohol intoxication and abstinence. Addiction Biology, 20(2), 263–274. 10.1111/adb.12119 [DOI] [PMC free article] [PubMed] [Google Scholar]
  202. Isherwood SN, Pekcec A, Nicholson JR, Robbins TW, & Dalley JW (2015). Dissociable effects of mGluR5 allosteric modulation on distinct forms of impulsivity in rats: interaction with NMDA receptor antagonism. Psychopharmacology, 232(18), 3327–3344. 10.1007/s00213-015-3984-0 [DOI] [PubMed] [Google Scholar]
  203. Isherwood SN, Robbins TW, Nicholson JR, Dalley JW, & Pekcec A (2017). Selective and interactive effects of D2 receptor antagonism and positive allosteric mGluR4 modulation on waiting impulsivity. Neuropharmacology, 123, 249–260. 10.1016/j.neuropharm.2017.05.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  204. Ito M (1984). The modifiable neuronal network of the cerebellum. The Japanese Journal of Physiology, 34(5), 781–792. 10.2170/jjphysiol.34.781 [DOI] [PubMed] [Google Scholar]
  205. Ito K, Abekawa T, & Koyama T (2006). Relationship between development of cross-sensitization to MK-801 and delayed increases in glutamate levels in the nucleus accumbens induced by a high dose of methamphetamine. Psychopharmacology, 187(3), 293–302. 10.1007/s00213-006-0423-2 [DOI] [PubMed] [Google Scholar]
  206. Jensen V, Rinholm JE, Johansen TJ, Medin T, Storm-Mathisen J, Sagvolden T, Hvalby O, & Bergersen LH (2009). N-methyl-D-aspartate receptor subunit dysfunction at hippocampal glutamatergic synapses in an animal model of attention-deficit/hyperactivity disorder. Neuroscience, 158(1), 353–364. 10.1016/j.neuroscience.2008.05.016 [DOI] [PubMed] [Google Scholar]
  207. Jentsch JD (2005). Impaired visuospatial divided attention in the spontaneously hypertensive rat. Behavioural Brain Research, 157(2), 323–330. 10.1016/j.bbr.2004.07.011 [DOI] [PubMed] [Google Scholar]
  208. Jentsch JD, Ashenhurst JR, Cervantes MC, Groman SM, James AS, & Pennington ZT (2014). Dissecting impulsivity and its relationships to drug addictions. Annals of the New York Academy of Sciences, 1327, 1–26. 10.1111/nyas.12388 [DOI] [PMC free article] [PubMed] [Google Scholar]
  209. Jentsch JD, & Anzivino LA (2004). A low dose of the alpha2 agonist clonidine ameliorates the visual attention and spatial working memory deficits produced by phencyclidine administration to rats. Psychopharmacology, 175(1), 76–83. 10.1007/s00213-004-1772-3 [DOI] [PubMed] [Google Scholar]
  210. Jo S, Kim KU, Lee D, & Jung MW (2013). Effect of orbitofrontal cortex lesions on temporal discounting in rats. Behavioural Brain Research, 245, 22–28. 10.1016/j.bbr.2013.02.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  211. Jueptner M, & Weiller C (1998). A review of differences between basal ganglia and cerebellar control of movements as revealed by functional imaging studies. Brain, 121 (Pt 8), 1437–1449. 10.1093/brain/121.8.1437 [DOI] [PubMed] [Google Scholar]
  212. Jun C, Choi Y, Lim SM, Bae S, Hong YS, Kim JE, & Lyoo IK (2014). Disturbance of the glutamatergic system in mood disorders. Experimental Neurobiology, 23(1), 28–35. 10.5607/en.2014.23.1.28 [DOI] [PMC free article] [PubMed] [Google Scholar]
  213. Kaartinen M, Puura K, Helminen M, Salmelin R, Pelkonen E, & Juujärvi P (2014). Reactive aggression among children with and without autism spectrum disorder. Journal of Autism and Developmental Disorders, 44(10), 2383–2391. 10.1007/s10803-012-1743-1 [DOI] [PubMed] [Google Scholar]
  214. Kabbani D, Akika R, Wahid A, Daly AK, Cascorbi I, & Zgheib NK (2023). Pharmacogenomics in practice: a review and implementation guide. Frontiers in Pharmacology, 14, 1189976. 10.3389/fphar.2023.1189976 [DOI] [PMC free article] [PubMed] [Google Scholar]
  215. Kahl KG, Atalay S, Maudsley AA, Sheriff S, Cummings A, Frieling H, Schmitz B, Lanfermann H, & Ding XQ (2020). Altered neurometabolism in major depressive disorder: a whole brain 1H-magnetic resonance spectroscopic imaging study at 3T. Progress in Neuro-Psychopharmacology & Biological Psychiatry, 101, 109916. 10.1016/j.pnpbp.2020.109916 [DOI] [PubMed] [Google Scholar]
  216. Kahl CK, Swansburg R, Hai T, Wrightson JG, Bell T, Lemay JF, Kirton A, & MacMaster FP (2022). Differences in neurometabolites and transcranial magnetic stimulation motor maps in children with attention-deficit/hyperactivity disorder. Journal of Psychiatry & Neuroscience, 47(4), E239–E249. 10.1503/jpn.210186 [DOI] [PMC free article] [PubMed] [Google Scholar]
  217. Kaizuka T, & Takumi T (2018). Postsynaptic density proteins and their involvement in neurodevelopmental disorders. Journal of Biochemistry, 163(6), 447–455. 10.1093/jb/mvy022 [DOI] [PubMed] [Google Scholar]
  218. Kalivas PW (2009). The glutamate homeostasis hypothesis of addiction. Nature Reviews Neuroscience, 10(8), 561–572. 10.1038/nrn2515 [DOI] [PubMed] [Google Scholar]
  219. Kalloniatis M, Marc RE, & Murry RF (1996). Amino acid signatures in the primate retina. The Journal of Neuroscience, 16(21), 6807–6829. 10.1523/JNEUROSCI.16-21-06807.1996 [DOI] [PMC free article] [PubMed] [Google Scholar]
  220. Kandaswamy R, McQuillin A, Curtis D, & Gurling H (2014). Allelic association, DNA resequencing and copy number variation at the metabotropic glutamate receptor GRM7 gene locus in bipolar disorder. American Journal of Medical Genetics. Part B, Neuropsychiatric Genetics, 165B(4), 365–372. 10.1002/ajmg.b.32239 [DOI] [PMC free article] [PubMed] [Google Scholar]
  221. Kapur S, & Seeman P (2002). NMDA receptor antagonists ketamine and PCP have direct effects on the dopamine D(2) and serotonin 5-HT(2)receptors-implications for models of schizophrenia. Molecular Psychiatry, 7(8), 837–844. 10.1038/sj.mp.4001093 [DOI] [PubMed] [Google Scholar]
  222. Kaut O, Schmitt I, Hofmann A, Hoffmann P, Schlaepfer TE, Wüllner U, & Hurlemann R (2015). Aberrant NMDA receptor DNA methylation detected by epigenome-wide analysis of hippocampus and prefrontal cortex in major depression. European Archives of Psychiatry and Clinical Neuroscience, 265(4), 331–341. 10.1007/s00406-014-0572-y [DOI] [PubMed] [Google Scholar]
  223. Kavoussi RJ, & Coccaro EF (1998). Divalproex sodium for impulsive aggressive behavior in patients with personality disorder. The Journal of Clinical Psychiatry, 59(12), 676–680. 10.4088/jcp.v59n1206 [DOI] [PubMed] [Google Scholar]
  224. Kheramin S, Body S, Mobini S, Ho MY, Velázquez-Martinez DN, Bradshaw CM, Szabadi E, Deakin JF, & Anderson IM (2002). Effects of quinolinic acid-induced lesions of the orbital prefrontal cortex on inter-temporal choice: a quantitative analysis. Psychopharmacology, 165(1), 9–17. 10.1007/s00213-002-1228-6 [DOI] [PubMed] [Google Scholar]
  225. Khurana A, Romer D, Betancourt LM, & Hurt H (2017). Working memory ability and early drug use progression as predictors of adolescent substance use disorders. Addiction, 112(7), 1220–1228. 10.1111/add.13792 [DOI] [PMC free article] [PubMed] [Google Scholar]
  226. Kilonzo K, Strahnen D, Prex V, Gems J, van der Veen B, Kapanaiah SKT, Murthy BKB, Schulz S, Sprengel R, Bannerman D, & Kätzel D (2022). Distinct contributions of GluA1-containing AMPA receptors of different hippocampal subfields to salience processing, memory and impulse control. Translational Psychiatry, 12(1), 102. 10.1038/s41398-022-01863-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  227. Kim DJ, Bartlett EA, DeLorenzo C, Parsey RV, Kilts C, & Cáceda R (2021). Examination of structural brain changes in recent suicidal behavior. Psychiatry Research. Neuroimaging, 307, 111216. 10.1016/j.pscychresns.2020.111216 [DOI] [PMC free article] [PubMed] [Google Scholar]
  228. Kim MJ, Dunah AW, Wang YT, & Sheng M (2005). Differential roles of NR2A- and NR2B-containing NMDA receptors in Ras-ERK signaling and AMPA receptor trafficking. Neuron, 46(5), 745–760. 10.1016/j.neuron.2005.04.031 [DOI] [PubMed] [Google Scholar]
  229. Kim S, & Lee D (2011). Prefrontal cortex and impulsive decision making. Biological Psychiatry, 69(12), 1140–1146. 10.1016/j.biopsych.2010.07.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  230. Kim JI, Kim JW, Park JE, Park S, Hong SB, Han DH, Cheong JH, Choi JW, Lee S, & Kim BN (2017). Association of the GRIN2B rs2284411 polymorphism with methylphenidate response in attention-deficit/hyperactivity disorder. Journal of Psychopharmacology, 31(8), 1070–1077. 10.1177/0269881116667707 [DOI] [PubMed] [Google Scholar]
  231. Kim E, Naisbitt S, Hsueh YP, Rao A, Rothschild A, Craig AM, & Sheng M (1997). GKAP, a novel synaptic protein that interacts with the guanylate kinase-like domain of the PSD-95/SAP90 family of channel clustering molecules. The Journal of Cell Biology, 136(3), 669–678. 10.1083/jcb.136.3.669 [DOI] [PMC free article] [PubMed] [Google Scholar]
  232. Kim JI, Yoo JH, Kim D, Jeong B, & Kim BN (2018). The effects of GRIN2B and DRD4 gene variants on local functional connectivity in attention-deficit/hyperactivity disorder. Brain Imaging and Behavior, 12(1), 247–257. 10.1007/s11682-017-9690-2 [DOI] [PubMed] [Google Scholar]
  233. Klein AB, Ultved L, Adamsen D, Santini MA, Tobeña A, Fernandez-Teruel A, Flores P, Moreno M, Cardona D, Knudsen GM, Aznar S, & Mikkelsen JD (2014). 5-HT(2A) and mGlu2 receptor binding levels are related to differences in impulsive behavior in the Roman Low- (RLA) and High- (RHA) avoidance rat strains. Neuroscience, 263, 36–45. 10.1016/j.neuroscience.2013.12.063 [DOI] [PubMed] [Google Scholar]
  234. Koizumi A, Lau H, Shimada Y, & Kondo HM (2018). The effects of neurochemical balance in the anterior cingulate cortex and dorsolateral prefrontal cortex on volitional control under irrelevant distraction. Consciousness and Cognition, 59, 104–111. 10.1016/j.concog.2018.01.001 [DOI] [PubMed] [Google Scholar]
  235. Kruse AO, & Bustillo JR (2022). Glutamatergic dysfunction in schizophrenia. Translational Psychiatry, 12(1), 500. 10.1038/s41398-022-02253-w [DOI] [PMC free article] [PubMed] [Google Scholar]
  236. Kumar M, Tripathi CD, Verma V, Padhy BM, Meshram GG, & Abhilash B (2016). Predictive validity of some common animal models of bipolar disorder using lithium and lamotrigine therapy: an attempt towards a battery-based approach for the evaluation of mood stabilizers. Psychiatry Investigation, 13(4), 434–439. 10.4306/pi.2016.13.4.434 [DOI] [PMC free article] [PubMed] [Google Scholar]
  237. Lan MJ, McLoughlin GA, Griffin JL, Tsang TM, Huang JT, Yuan P, Manji H, Holmes E, & Bahn S (2009). Metabonomic analysis identifies molecular changes associated with the pathophysiology and drug treatment of bipolar disorder. Molecular Psychiatry, 14(3), 269–279. 10.1038/sj.mp.4002130 [DOI] [PubMed] [Google Scholar]
  238. Laurin N, Lee J, Ickowicz A, Pathare T, Malone M, Tannock R, Kennedy JL, Schachar RJ, & Barr CL (2008). Association study for genes at chromosome 5p13-q11 in attention deficit hyperactivity disorder. American Journal of Medical Genetics. Part B, Neuropsychiatric Genetics, 147B(5), 600–605. 10.1002/ajmg.b.30654 [DOI] [PubMed] [Google Scholar]
  239. Leclerc MP, Regenbogen C, Hamilton RH, & Habel U (2018). Some neuroanatomical insights to impulsive aggression in schizophrenia. Schizophrenia Research, 201, 27–34. 10.1016/j.schres.2018.06.016 [DOI] [PubMed] [Google Scholar]
  240. Lee RSC, Hoppenbrouwers S, & Franken I (2019). A systematic meta-review of impulsivity and compulsivity in addictive behaviors. Neuropsychology Review, 29(1), 14–26. 10.1007/s11065-019-09402-x [DOI] [PubMed] [Google Scholar]
  241. Lee G, & Zhou Y (2019). NMDAR hypofunction animal models of schizophrenia. Frontiers in Molecular Neuroscience, 12, 185. 10.3389/fnmol.2019.00185 [DOI] [PMC free article] [PubMed] [Google Scholar]
  242. Leffa DT, Panzenhagen AC, Salvi AA, Bau CHD, Pires GN, Torres ILS, Rohde LA, Rovaris DL, & Grevet EH (2019). Systematic review and meta-analysis of the behavioral effects of methylphenidate in the spontaneously hypertensive rat model of attention-deficit/hyperactivity disorder. Neuroscience and Biobehavioral Reviews, 100, 166–179. 10.1016/j.neubiorev.2019.02.019 [DOI] [PubMed] [Google Scholar]
  243. Lehohla M, Kellaway L, & Russell VA (2004). NMDA receptor function in the prefrontal cortex of a rat model for attention-deficit hyperactivity disorder. Metabolic Brain Disease, 19(1–2), 35–42. 10.1023/b:mebr.0000027415.75432.ad [DOI] [PubMed] [Google Scholar]
  244. Léna I, Chessel A, Le Pen G, Krebs MO, & Garcia R (2007). Alterations in prefrontal glutamatergic and noradrenergic systems following MK-801 administration in rats prenatally exposed to methylazoxymethanol at gestational day 17. Psychopharmacology, 192(3), 373–383. 10.1007/s00213-007-0719-x [DOI] [PubMed] [Google Scholar]
  245. Lesch KP, Merker S, Reif A, & Novak M (2013). Dances with black widow spiders: dysregulation of glutamate signalling enters centre stage in ADHD. European Neuropsychopharmacology, 23(6), 479–491. 10.1016/j.euroneuro.2012.07.013 [DOI] [PubMed] [Google Scholar]
  246. Levine J, Panchalingam K, Rapoport A, Gershon S, McClure RJ, & Pettegrew JW (2000). Increased cerebrospinal fluid glutamine levels in depressed patients. Biological Psychiatry, 47(7), 586–593. 10.1016/s0006-3223(99)00284-x [DOI] [PubMed] [Google Scholar]
  247. Levran O, Londono D, O’Hara K, Randesi M, Rotrosen J, Casadonte P, Linzy S, Ott J, Adelson M, & Kreek MJ (2009). Heroin addiction in African Americans: a hypothesis-driven association study. Genes, Brain, and Behavior, 8(5), 531–540. 10.1111/j.1601-183X.2009.00501.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  248. Li H, Chen J, Chen C, Xu Z, Xu J, Lin W, Wu J, Li G, Xu H, Kang Y, Wang F, & Liu Y (2018). CSF glutamate level decreases in heavy smokers and negatively correlates with BDI scores. Psychiatry Research, 270, 627–630. 10.1016/j.psychres.2018.10.053 [DOI] [PubMed] [Google Scholar]
  249. Li JN, Liu XL, & Li L (2020). Prefrontal GABA and glutamate levels correlate with impulsivity and cognitive function of prescription opioid addicts: A 1 H-magnetic resonance spectroscopy study. Psychiatry and Clinical Neurosciences, 74(1), 77–83. 10.1111/pcn.12940 [DOI] [PubMed] [Google Scholar]
  250. Li B, Zhang S, Li M, Zhang H, Hertz L, & Peng L (2009). Down-regulation of GluK2 kainate receptor expression by chronic treatment with mood-stabilizing anti-convulsants or lithium in cultured astrocytes and brain, but not in neurons. Neuropharmacology, 57(4), 375–385. 10.1016/j.neuropharm.2009.07.004 [DOI] [PubMed] [Google Scholar]
  251. Lin CH, Huang MW, Lin CH, Huang CH, & Lane HY (2019). Altered mRNA expressions for N-methyl-D-aspartate receptor-related genes in WBC of patients with major depressive disorder. Journal of Affective Disorders, 245, 1119–1125. 10.1016/j.jad.2018.12.016 [DOI] [PubMed] [Google Scholar]
  252. Lin Z, Lu Y, Li S, Li Y, Li H, Li L, & Wang L (2022). Effect of eplerenone on cognitive impairment in spontaneously hypertensive rats. American Journal of Translational Research, 14(6), 3864–3878. [PMC free article] [PubMed] [Google Scholar]
  253. Lisek M, Ferenc B, Studzian M, Pulaski L, Guo F, Zylinska L, & Boczek T (2017). Glutamate deregulation in ketamine-induced psychosis-a potential role of PSD95, NMDA receptor and PMCA interaction. Frontiers in Cellular Neuroscience, 11, 181. 10.3389/fncel.2017.00181 [DOI] [PMC free article] [PubMed] [Google Scholar]
  254. Liu P, & Bilkey DK (1996). Direct connection between perirhinal cortex and hippocampus is a major constituent of the lateral perforant path. Hippocampus, 6(2), 125–135. [DOI] [PubMed] [Google Scholar]
  255. Liu F, Grauer S, Kelley C, Navarra R, Graf R, Zhang G, Atkinson PJ, Popiolek M, Wantuch C, Khawaja X, Smith D, Olsen M, Kouranova E, Lai M, Pruthi F, Pulicicchio C, Day M, Gilbert A, Pausch MH, Brandon NJ, … Marquis KL (2008). ADX47273 [S-(4-fluoro-phenyl)-{3-[3-(4-fluoro-phenyl)-[1,2,4]-oxadiazol-5-yl]-piperidin-1-yl}-methanone]: a novel metabotropic glutamate receptor 5-selective positive allosteric modulator with preclinical antipsychotic-like and procognitive activities. The Journal of Pharmacology and Experimental Therapeutics, 327(3), 827–839. 10.1124/jpet.108.136580 [DOI] [PubMed] [Google Scholar]
  256. Liu SJ, Lan Y, Wu L, & Yan WS (2019). Profiles of impulsivity in problematic internet users and cigarette smokers. Frontiers in Psychology, 10, 772. 10.3389/fpsyg.2019.00772 [DOI] [PMC free article] [PubMed] [Google Scholar]
  257. Liu Y, Qu HQ, Chang X, Nguyen K, Qu J, Tian L, Glessner J, Sleiman PM, & Hakonarson H (2021). Deep learning prediction of attention-deficit hyperactivity disorder in African Americans by copy number variation. Experimental Biology and Medicine, 246(21), 2317–2323. 10.1177/15353702211018970 [DOI] [PMC free article] [PubMed] [Google Scholar]
  258. Livesey PJ, Meyer P, & Smith JP (1980). Effects of disruption of hippocampal function on learning of a go/no-go discrimination in the rat. Brain Research, 195(1), 197–202. 10.1016/0006-8993(80)90877-x [DOI] [PubMed] [Google Scholar]
  259. Logan GD, & Cowan WB (1984). On the ability to inhibit thought and action: a theory of an act of control. Psychological Review, 91(3), 295–327. 10.1037/0033-295X.91.3.295 [DOI] [PubMed] [Google Scholar]
  260. Logan GD, Cowan WB, & Davis KA (1984). On the ability to inhibit simple and choice reaction time responses: a model and a method. Journal of Experimental Psychology. Human Perception and Performance, 10(2), 276–291. 10.1037//0096-1523.10.2.276 [DOI] [PubMed] [Google Scholar]
  261. Lorenz RC, Gleich T, Buchert R, Schlagenhauf F, Kühn S, & Gallinat J (2015). Interactions between glutamate, dopamine, and the neuronal signature of response inhibition in the human striatum. Human Brain Mapping, 36(10), 4031–4040. 10.1002/hbm.22895 [DOI] [PMC free article] [PubMed] [Google Scholar]
  262. Luo SX, & Huang EJ (2016). Dopaminergic neurons and brain reward pathways: from neurogenesis to circuit assembly. The American Journal of Pathology, 186(3), 478–488. 10.1016/j.ajpath.2015.09.023 [DOI] [PMC free article] [PubMed] [Google Scholar]
  263. Luo P, Li A, Zheng Y, Han Y, Tian J, Xu Z, Gong H, & Li X (2019). Whole brain mapping of long-range direct input to glutamatergic and GABAergic neurons in motor cortex. Frontiers in Neuroanatomy, 13, 44. 10.3389/fnana.2019.00044 [DOI] [PMC free article] [PubMed] [Google Scholar]
  264. Lüscher C, & Huber KM (2010). Group 1 mGluR-dependent synaptic long-term depression: mechanisms and implications for circuitry and disease. Neuron, 65(4), 445–459. 10.1016/j.neuron.2010.01.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  265. Lüscher C, & Malenka RC (2012). NMDA receptor-dependent long-term potentiation and long-term depression (LTP/LTD). Cold Spring Harbor Perspectives in Biology, 4(6), a005710. 10.1101/cshperspect.a005710 [DOI] [PMC free article] [PubMed] [Google Scholar]
  266. Ma H, Xun G, Zhang R, Yang X, & Cao Y (2019). Correlation between GRIK2 rs6922753, rs2227283 polymorphism and aggressive behaviors with Bipolar Mania in the Chinese Han population. Brain and Behavior, 9(11), e01449. 10.1002/brb3.1449 [DOI] [PMC free article] [PubMed] [Google Scholar]
  267. MacMaster FP, Carrey N, Sparkes S, & Kusumakar V (2003). Proton spectroscopy in medication-free pediatric attention-deficit/hyperactivity disorder. Biological Psychiatry, 53(2), 184–187. 10.1016/s0006-3223(02)01401-4 [DOI] [PubMed] [Google Scholar]
  268. MacMillan CM, Korndörfer SR, Rao S, Fleisher CA, Mezzacappa E, & Gonzalez-Heydrich J (2006). A comparison of divalproex and oxcarbazepine in aggressive youth with bipolar disorder. Journal of Psychiatric Practice, 12(4), 214–222. 10.1097/00131746-200607000-00003 [DOI] [PubMed] [Google Scholar]
  269. Madden GJ, & Johnson PS (2010). A delay-discounting primer. In Madden GJ & Bickel WK (Eds.), Impulsivity: The behavioral and neurological science of discounting (pp. 11–37). American Psychological Association. 10.1037/12069-001 [DOI] [Google Scholar]
  270. Madison DV, Malenka RC, & Nicoll RA (1991). Mechanisms underlying long-term potentiation of synaptic transmission. Annual Review of Neuroscience, 14, 379–397. 10.1146/annurev.ne.14.030191.002115 [DOI] [PubMed] [Google Scholar]
  271. Makinson R, Lloyd K, Grissom N, & Reyes TM (2019). Exposure to in utero inflammation increases locomotor activity, alters cognitive performance and drives vulnerability to cognitive performance deficits after acute immune activation. Brain, Behavior, and Immunity, 80, 56–65. 10.1016/j.bbi.2019.02.022 [DOI] [PubMed] [Google Scholar]
  272. Malenka RC, & Bear MF (2004). LTP and LTD: an embarrassment of riches. Neuron, 44(1), 5–21. 10.1016/j.neuron.2004.09.012 [DOI] [PubMed] [Google Scholar]
  273. Maltezos S, Horder J, Coghlan S, Skirrow C, O’Gorman R, Lavender TJ, Mendez MA, Mehta M, Daly E, Xenitidis K, Paliokosta E, Spain D, Pitts M, Asherson P, Lythgoe DJ, Barker GJ, & Murphy DG (2014). Glutamate/glutamine and neuronal integrity in adults with ADHD: a proton MRS study. Translational Psychiatry, 4(3), e373. 10.1038/tp.2014.11 [DOI] [PMC free article] [PubMed] [Google Scholar]
  274. Mandal S, Sinha VK, & Goyal N (2019). Efficacy of ketamine therapy in the treatment of depression. Indian Journal of Psychiatry, 61(5), 480–485. 10.4103/psychiatry.IndianJPsychiatry_484_18 [DOI] [PMC free article] [PubMed] [Google Scholar]
  275. Mann JJ, Arango V, & Underwood MD (1990). Serotonin and suicidal behavior. Annals of the New York Academy of Sciences, 600, 476–485. 10.1111/j.1749-6632.1990.tb16903.x [DOI] [PubMed] [Google Scholar]
  276. Mann LG, Hay KR, Song AK, Errington SP, Trujillo P, Zald DH, Yan Y, Kang H, Logan GD, & Claassen DO (2021). D2-like receptor expression in the hippocampus and amygdala informs performance on the stop-signal task in Parkinson’s disease. The Journal of Neuroscience, 41(48), 10023–10030. 10.1523/JNEUROSCI.0968-21.2021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  277. Mar AC, Walker AL, Theobald DE, Eagle DM, & Robbins TW (2011). Dissociable effects of lesions to orbitofrontal cortex subregions on impulsive choice in the rat. The Journal of Neuroscience, 31(17), 6398–6404. 10.1523/JNEUROSCI.6620-10.2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  278. Mariano TY, Bannerman DM, McHugh SB, Preston TJ, Rudebeck PH, Rudebeck SR, Rawlins JN, Walton ME, Rushworth MF, Baxter MG, & Campbell TG (2009). Impulsive choice in hippocampal but not orbitofrontal cortex-lesioned rats on a nonspatial decision-making maze task. The European Journal of Neuroscience, 30(3), 472–484. 10.1111/j.1460-9568.2009.06837.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  279. Martucci L, Wong AH, De Luca V, Likhodi O, Wong GW, King N, & Kennedy JL (2006). N-methyl-D-aspartate receptor NR2B subunit gene GRIN2B in schizophrenia and bipolar disorder: polymorphisms and mRNA levels. Schizophrenia Research, 84(2–3), 214–221. 10.1016/j.schres.2006.02.001 [DOI] [PubMed] [Google Scholar]
  280. Marusich JA, Darna M, Charnigo RJ, Dwoskin LP, & Bardo MT (2011). A multivariate assessment of individual differences in sensation seeking and impulsivity as predictors of amphetamine self-administration and prefrontal dopamine function in rats. Experimental and Clinical Psychopharmacology, 19(4), 275–284. 10.1037/a0023897 [DOI] [PMC free article] [PubMed] [Google Scholar]
  281. Mazur JE (1987). An adjusting procedure for studying delayed reinforcement. In Commons ML, Mazur JE, Nevin JA, & Rachlin H (Eds.), The effect of delay and of intervening events on reinforcement value (pp. 55–73). Lawrence Erlbaum Associates, Inc. [Google Scholar]
  282. McOmish CE, Demireva EY, & Gingrich JA (2016). Developmental expression of mGlu2 and mGlu3 in the mouse brain. Gene Expression Patterns, 22(2), 46–53. 10.1016/j.gep.2016.10.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  283. Medin T, Jensen V, Skare Ø, Storm-Mathisen J, Hvalby Ø, & Bergersen LH (2019). Altered α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor function and expression in hippocampus in a rat model of attention-deficit/hyperactivity disorder (ADHD). Behavioural Brain Research, 360, 209–215. 10.1016/j.bbr.2018.12.028 [DOI] [PubMed] [Google Scholar]
  284. Medina A, Burke S, Thompson RC, Bunney W Jr, Myers RM, Schatzberg A, Akil H, & Watson SJ (2013). Glutamate transporters: a key piece in the glutamate puzzle of major depressive disorder. Journal of Psychiatric Research, 47(9), 1150–1156. 10.1016/j.jpsychires.2013.04.007 [DOI] [PubMed] [Google Scholar]
  285. Michael N, Erfurth A, & Pfleiderer B (2009). Elevated metabolites within dorsolateral prefrontal cortex in rapid cycling bipolar disorder. Psychiatry Research, 172(1), 78–81. 10.1016/j.pscychresns.2009.01.002 [DOI] [PubMed] [Google Scholar]
  286. Michelsen KA, Prickaerts J, & Steinbusch HW (2008). The dorsal raphe nucleus and serotonin: implications for neuroplasticity linked to major depression and Alzheimer’s disease. Progress in Brain Research, 172, 233–264. 10.1016/S0079-6123(08)00912-6 [DOI] [PubMed] [Google Scholar]
  287. Miller KE, Hoffman EM, Sutharshan M, & Schechter R (2011). Glutamate pharmacology and metabolism in peripheral primary afferents: physiological and pathophysiological mechanisms. Pharmacology & Therapeutics, 130(3), 283–309. 10.1016/j.pharmthera.2011.01.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  288. Miller EM, Pomerleau F, Huettl P, Gerhardt GA, & Glaser PE (2014). Aberrant glutamate signaling in the prefrontal cortex and striatum of the spontaneously hypertensive rat model of attention-deficit/hyperactivity disorder. Psychopharmacology, 231(15), 3019–3029. 10.1007/s00213-014-3479-4 [DOI] [PubMed] [Google Scholar]
  289. Miller EM, Quintero JE, Pomerleau F, Huettl P, Gerhardt GA, & Glaser PEA (2019). Chronic methylphenidate alters tonic and phasic glutamate signaling in the frontal cortex of a freely-moving rat model of ADHD. Neurochemical Research, 44(1), 89–101. 10.1007/s11064-018-2483-1 [DOI] [PubMed] [Google Scholar]
  290. Mitchell SH, Reeves JM, Li N, & Phillips TJ (2006). Delay discounting predicts behavioral sensitization to ethanol in outbred WSC mice. Alcoholism: Clinical and Experimental Research, 30(3), 429–437. 10.1111/j.1530-0277.2006.00047.x [DOI] [PubMed] [Google Scholar]
  291. Mobini S, Body S, Ho MY, Bradshaw CM, Szabadi E, Deakin JF, & Anderson IM (2002). Effects of lesions of the orbitofrontal cortex on sensitivity to delayed and probabilistic reinforcement. Psychopharmacology, 160(3), 290–298. 10.1007/s00213-001-0983-0 [DOI] [PubMed] [Google Scholar]
  292. Moghaddam B, Adams B, Verma A, & Daly D (1997). Activation of glutamatergic neurotransmission by ketamine: a novel step in the pathway from NMDA receptor blockade to dopaminergic and cognitive disruptions associated with the prefrontal cortex. The Journal of Neuroscience, 17(8), 2921–2927. 10.1523/JNEUROSCI.17-08-02921.1997 [DOI] [PMC free article] [PubMed] [Google Scholar]
  293. Moghaddam B, & Javitt D (2012). From revolution to evolution: the glutamate hypothesis of schizophrenia and its implication for treatment. Neuropsychopharmacology, 37(1), 4–15. 10.1038/npp.2011.181 [DOI] [PMC free article] [PubMed] [Google Scholar]
  294. Monaghan DT, & Cotman CW (1985). Distribution of N-methyl-D-aspartate-sensitive L-[3H]glutamate-binding sites in rat brain. The Journal of Neuroscience, 5(11), 2909–2919. 10.1523/JNEUROSCI.05-11-02909.1985 [DOI] [PMC free article] [PubMed] [Google Scholar]
  295. Monaghan DT, Yao D, Olverman HJ, Watkins JC, & Cotman CW (1984). Autoradiography of D-2-[3H]amino-5-phosphonopentanoate binding sites in rat brain. Neuroscience Letters, 52(3), 253–258. 10.1016/0304-3940(84)90170-8 [DOI] [PubMed] [Google Scholar]
  296. Monteiro P, & Feng G (2017). SHANK proteins: roles at the synapse and in autism spectrum disorder. Nature Reviews. Neuroscience, 18(3), 147–157. 10.1038/nrn.2016.183 [DOI] [PubMed] [Google Scholar]
  297. Morgane PJ, Galler JR, & Mokler DJ (2005). A review of systems and networks of the limbic forebrain/limbic midbrain. Progress in Neurobiology, 75(2), 143–160. 10.1016/j.pneurobio.2005.01.001 [DOI] [PubMed] [Google Scholar]
  298. Morigaki R, Miyamoto R, Matsuda T, Miyake K, Yamamoto N, & Takagi Y (2021). Dystonia and cerebellum: from bench to bedside. Life, 11(8), 776. 10.3390/life11080776 [DOI] [PMC free article] [PubMed] [Google Scholar]
  299. Morishima Y, Miyakawa T, Furuyashiki T, Tanaka Y, Mizuma H, & Nakanishi S (2005). Enhanced cocaine responsiveness and impaired motor coordination in metabotropic glutamate receptor subtype 2 knockout mice. Proceedings of the National Academy of Sciences of the United States of America, 102(11), 4170–4175. 10.1073/pnas.0500914102 [DOI] [PMC free article] [PubMed] [Google Scholar]
  300. Murphy ER, Dalley JW, & Robbins TW (2005). Local glutamate receptor antagonism in the rat prefrontal cortex disrupts response inhibition in a visuospatial attentional task. Psychopharmacology, 179(1), 99–107. 10.1007/s00213-004-2068-3 [DOI] [PubMed] [Google Scholar]
  301. Murphy ER, Fernando AB, Urcelay GP, Robinson ES, Mar AC, Theobald DE, Dalley JW, & Robbins TW (2012). Impulsive behaviour induced by both NMDA receptor antagonism and GABAA receptor activation in rat ventromedial prefrontal cortex. Psychopharmacology, 219(2), 401–410. 10.1007/s00213-011-2572-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  302. Murphy GJ, Glickfeld LL, Balsen Z, & Isaacson JS (2004). Sensory neuron signaling to the brain: properties of transmitter release from olfactory nerve terminals. The Journal of Neuroscience, 24(12), 3023–3030. 10.1523/JNEUROSCI.5745-03.2004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  303. Murrough JW, Iosifescu DV, Chang LC, Al Jurdi RK, Green CE, Perez AM, Iqbal S, Pillemer S, Foulkes A, Shah A, Charney DS, & Mathew SJ (2013). Antidepressant efficacy of ketamine in treatment-resistant major depression: a two-site randomized controlled trial. The American Journal of Psychiatry, 170(10), 1134–1142. 10.1176/appi.ajp.2013.13030392 [DOI] [PMC free article] [PubMed] [Google Scholar]
  304. Murrough JW, Soleimani L, DeWilde KE, Collins KA, Lapidus KA, Iacoviello BM, Lener M, Kautz M, Kim J, Stern JB, Price RB, Perez AM, Brallier JW, Rodriguez GJ, Goodman WK, Iosifescu DV, & Charney DS (2015). Ketamine for rapid reduction of suicidal ideation: a randomized controlled trial. Psychological Medicine, 45(16), 3571–3580. 10.1017/S0033291715001506 [DOI] [PubMed] [Google Scholar]
  305. Naaijen J, Forde NJ, Lythgoe DJ, Akkermans SE, Openneer TJ, Dietrich A, Zwiers MP, Hoekstra PJ, & Buitelaar JK (2016). Fronto-striatal glutamate in children with Tourette’s disorder and attention-deficit/hyperactivity disorder. NeuroImage. Clinical, 13, 16–23. 10.1016/j.nicl.2016.11.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  306. Nambu A, Tokuno H, & Takada M (2002). Functional significance of the cortico-subthalamo-pallidal ‘hyperdirect’ pathway. Neuroscience Research, 43(2), 111–117. 10.1016/s0168-0102(02)00027-5 [DOI] [PubMed] [Google Scholar]
  307. Nachev P, Kennard C, & Husain M (2008). Functional role of the supplementary and pre-supplementary motor areas. Nature Reviews. Neuroscience, 9(11), 856–869. 10.1038/nrn2478 [DOI] [PubMed] [Google Scholar]
  308. Nejati V, Sarraj Khorrami A, & Nitsche MA (2021). Transcranial direct current stimulation improves reward processing in children with ADHD. Journal of Attention Disorders, 25(11), 1623–1631. 10.1177/1087054720923094 [DOI] [PubMed] [Google Scholar]
  309. Newman JP, Widom CS, & Nathan S (1985). Passive avoidance in syndromes of disinhibition: psychopathy and extraversion. Journal of Personality and Social Psychology, 48(5), 1316–1327. 10.1037//0022-3514.48.5.1316 [DOI] [PubMed] [Google Scholar]
  310. Nicholls DG, & Sihra TS (1986). Synaptosomes possess an exocytotic pool of glutamate. Nature, 321(6072), 772–773. 10.1038/321772a0 [DOI] [PubMed] [Google Scholar]
  311. Nisar S, Bhat AA, Masoodi T, Hashem S, Akhtar S, Ali TA, Amjad S, Chawla S, Bagga P, Frenneaux MP, Reddy R, Fakhro K, & Haris M (2022). Genetics of glutamate and its receptors in autism spectrum disorder. Molecular Psychiatry, 27(5), 2380–2392. 10.1038/s41380-022-01506-w [DOI] [PMC free article] [PubMed] [Google Scholar]
  312. Nishi D, Hashimoto K, Noguchi H, Hamazaki K, Hamazaki T, & Matsuoka Y (2015). Glutamatergic system abnormalities in posttraumatic stress disorder. Psychopharmacology, 232(23), 4261–4268. 10.1007/s00213-015-4052-5 [DOI] [PubMed] [Google Scholar]
  313. Niswender CM, & Conn PJ (2010). Metabotropic glutamate receptors: physiology, pharmacology, and disease. Annual Review of Pharmacology and Toxicology, 50, 295–322. 10.1146/annurev.pharmtox.011008.145533 [DOI] [PMC free article] [PubMed] [Google Scholar]
  314. Nord CL, Kim SG, Callesen MB, Kvamme TL, Jensen M, Pedersen MU, Thomsen KR, & Voon V (2019). The myeloarchitecture of impulsivity: premature responding in youth is associated with decreased myelination of ventral putamen. Neuropsychopharmacology, 44(7), 1216–1223. 10.1038/s41386-019-0343-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  315. Noroozi R, Taheri M, Omrani MD, & Ghafouri-Fard S (2019). Glutamate receptor metabotropic 7 (GRM7) gene polymorphisms in mood disorders and attention deficit hyperactive disorder. Neurochemistry International, 129, 104483. 10.1016/j.neuint.2019.104483 [DOI] [PubMed] [Google Scholar]
  316. Nudmamud-Thanoi S, & Reynolds GP (2004). The NR1 subunit of the glutamate/NMDA receptor in the superior temporal cortex in schizophrenia and affective disorders. Neuroscience Letters, 372(1–2), 173–177. 10.1016/j.neulet.2004.09.035 [DOI] [PubMed] [Google Scholar]
  317. Ogawa S, Koga N, Hattori K, Matsuo J, Ota M, Hori H, Sasayama D, Teraishi T, Ishida I, Yoshida F, Yoshida S, Noda T, Higuchi T, & Kunugi H (2018). Plasma amino acid profile in major depressive disorder: analyses in two independent case-control sample sets. Journal of Psychiatric Research, 96, 23–32. 10.1016/j.jpsychires.2017.09.014 [DOI] [PubMed] [Google Scholar]
  318. Ohbayashi M (2021). The roles of the cortical motor areas in sequential movements. Frontiers in Behavioral Neuroscience, 15, 640659. 10.3389/fnbeh.2021.640659 [DOI] [PMC free article] [PubMed] [Google Scholar]
  319. Ohishi H, Akazawa C, Shigemoto R, Nakanishi S, & Mizuno N (1995). Distributions of the mRNAs for L-2-amino-4-phosphonobutyrate-sensitive metabotropic glutamate receptors, mGluR4 and mGluR7, in the rat brain. The Journal of Comparative Neurology, 360(4), 555–570. 10.1002/cne.903600402 [DOI] [PubMed] [Google Scholar]
  320. Ohishi H, Neki A, & Mizuno N (1998). Distribution of a metabotropic glutamate receptor, mGluR2, in the central nervous system of the rat and mouse: an immunohistochemical study with a monoclonal antibody. Neuroscience Research, 30(1), 65–82. 10.1016/s0168-0102(97)00120-x [DOI] [PubMed] [Google Scholar]
  321. Olié E, Doell KC, Corradi-Dell’Acqua C, Courtet P, Perroud N, & Schwartz S (2018). Physical pain recruits the nucleus accumbens during social distress in borderline personality disorder. Social Cognitive and Affective Neuroscience, 13(10), 1071–1080. 10.1093/scan/nsy078 [DOI] [PMC free article] [PubMed] [Google Scholar]
  322. Olney JW (1986). Inciting excitotoxic cytocide among central neurons. Advances in Experimental Medicine and Biology, 203, 631–645. 10.1007/978-1-4684-7971-3_48 [DOI] [PubMed] [Google Scholar]
  323. Olsen O, & Bredt DS (2003). Functional analysis of the nucleotide binding domain of membrane-associated guanylate kinases. The Journal of Biological Chemistry, 278(9), 6873–6878. 10.1074/jbc.M210165200 [DOI] [PubMed] [Google Scholar]
  324. Onofrychuk TJ, Cai S, McElroy DL, Roebuck AJ, Greba Q, Garai S, Thakur GA, Laprairie RB, & Howland JG (2021). Effects of the cannabinoid receptor 1 positive allosteric modulator GAT211 and acute MK-801 on visual attention and impulsivity in rats assessed using the five-choice serial reaction time task. Progress in Neuro-Psychopharmacology & Biological Psychiatry, 109, 110235. 10.1016/j.pnpbp.2020.110235 [DOI] [PubMed] [Google Scholar]
  325. Ortiz N, Parsons A, Whelan R, Brennan K, Agan ML, O’Connell R, Bramham J, & Garavan H (2015). Decreased frontal, striatal and cerebellar activation in adults with ADHD during an adaptive delay discounting task. Acta Neurobiologiae Experimentalis, 75(3), 326–338. 10.55782/ane-2015-2038 [DOI] [PubMed] [Google Scholar]
  326. Ozawa S, Kamiya H, & Tsuzuki K (1998). Glutamate receptors in the mammalian central nervous system. Progress in Neurobiology, 54(5), 581–618. 10.1016/s0301-0082(97)00085-3 [DOI] [PubMed] [Google Scholar]
  327. Paik SH, Choi MR, Kwak SM, Bang SH, & Kim DJ (2018). Decreased serum glutamate levels in male adults with internet gaming disorder: a pilot study. Clinical Psychopharmacology and Neuroscience, 16(3), 276–281. 10.9758/cpn.2018.16.3.276 [DOI] [PMC free article] [PubMed] [Google Scholar]
  328. Paine TA, Cooke EK, & Lowes DC (2015). Effects of chronic inhibition of GABA synthesis on attention and impulse control. Pharmacology, Biochemistry, and Behavior, 135, 97–104. 10.1016/j.pbb.2015.05.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  329. Palomino A, González-Pinto A, Aldama A, González-Gómez C, Mosquera F, González-García G, & Matute C (2007). Decreased levels of plasma glutamate in patients with first-episode schizophrenia and bipolar disorder. Schizophrenia Research, 95(1–3), 174–178. 10.1016/j.schres.2007.06.012 [DOI] [PubMed] [Google Scholar]
  330. Pålsson E, Jakobsson J, Södersten K, Fujita Y, Sellgren C, Ekman CJ, Ågren H, Hashimoto K, & Landén M (2015). Markers of glutamate signaling in cerebrospinal fluid and serum from patients with bipolar disorder and healthy controls. European Neuropsychopharmacology, 25(1), 133–140. 10.1016/j.euroneuro.2014.11.001 [DOI] [PubMed] [Google Scholar]
  331. Paraskevopoulou F, Herman MA, & Rosenmund C (2019). Glutamatergic innervation onto striatal neurons potentiates GABAergic synaptic output. The Journal of Neuroscience, 39(23), 4448–4460. 10.1523/JNEUROSCI.2630-18.2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  332. Pardini DA, Raine A, Erickson K, & Loeber R (2014). Lower amygdala volume in men is associated with childhood aggression, early psychopathic traits, and future violence. Biological Psychiatry, 75(1), 73–80. 10.1016/j.biopsych.2013.04.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  333. Parisi S, Gunasekara R, Canale C, Hasta F, & Azizi H (2021). The role of the anterior cingulate cortex and insular cortex in suicidal memory and intent. Cureus, 13(7), e16335. 10.7759/cureus.16335 [DOI] [PMC free article] [PubMed] [Google Scholar]
  334. Park S, Jung SW, Kim BN, Cho SC, Shin MS, Kim JW, Yoo HJ, Cho DY, Chung US, Son JW, & Kim HW (2013). Association between the GRM7 rs3792452 polymorphism and attention deficit hyperactivity disorder in a Korean sample. Behavioral and Brain Functions, 9, 1. 10.1186/1744-9081-9-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  335. Park S, Kim BN, Cho SC, Kim JW, Kim JI, Shin MS, Yoo HJ, Han DH, & Cheong JH (2014). The metabotropic glutamate receptor subtype 7 rs3792452 polymorphism is associated with the response to methylphenidate in children with attention-deficit/hyperactivity disorder. Journal of Child and Adolescent Psychopharmacology, 24(4), 223–227. 10.1089/cap.2013.0079 [DOI] [PubMed] [Google Scholar]
  336. Park CHK, Lee JW, Lee SY, Moon J, Jeon DW, Shim SH, Cho SJ, Kim SG, Lee J, Paik JW, Kim MH, You S, Jeon HJ, Rhee SJ, Kim MJ, Kim J, & Ahn YM (2020). Suicide risk factors across suicidal ideators, single suicide attempters, and multiple suicide attempters. Journal of Psychiatric Research, 131, 1–8. 10.1016/j.jpsychires.2020.08.018 [DOI] [PubMed] [Google Scholar]
  337. Pattij T, & Vanderschuren LJ (2008). The neuropharmacology of impulsive behaviour. Trends in Pharmacological Sciences, 29(4), 192–199. 10.1016/j.tips.2008.01.002 [DOI] [PubMed] [Google Scholar]
  338. Patton JH, Stanford MS, & Barratt ES (1995). Factor structure of the Barratt impulsiveness scale. Journal of Clinical Psychology, 51(6), 768–774. [DOI] [PubMed] [Google Scholar]
  339. Pawliczek CM, Derntl B, Kellermann T, Kohn N, Gur RC, & Habel U (2013). Inhibitory control and trait aggression: neural and behavioral insights using the emotional stop signal task. NeuroImage, 79, 264–274. 10.1016/j.neuroimage.2013.04.104 [DOI] [PubMed] [Google Scholar]
  340. Peng SX, Wang YY, Zhang M, Zang YY, Wu D, Pei J, Li Y, Dai J, Guo X, Luo X, Zhang N, Yang JJ, Zhang C, Gao X, Liu N, & Shi YS (2021). SNP rs10420324 in the AMPA receptor auxiliary subunit TARP γ-8 regulates the susceptibility to antisocial personality disorder. Scientific Reports, 11(1), 11997. 10.1038/s41598-021-91415-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  341. Perlov E, Tebarzt van Elst L, Buechert M, Maier S, Matthies S, Ebert D, Hesslinger B, & Philipsen A (2010). H1-MR-spectroscopy of cerebellum in adult attention deficit/hyperactivity disorder. Journal of Psychiatric Research, 44(14), 938–943. 10.1016/j.jpsychires.2010.02.016 [DOI] [PubMed] [Google Scholar]
  342. Perry JL, Larson EB, German JP, Madden GJ, & Carroll ME (2005). Impulsivity (delay discounting) as a predictor of acquisition of IV cocaine self-administration in female rats. Psychopharmacology, 178(2–3), 193–201. 10.1007/s00213-004-1994-4 [DOI] [PubMed] [Google Scholar]
  343. Peters J, & D’Esposito M (2016). Effects of medial orbitofrontal cortex lesions on self-control in intertemporal choice. Current Biology, 26(19), 2625–2628. 10.1016/j.cub.2016.07.035 [DOI] [PubMed] [Google Scholar]
  344. Pettorruso M, De Risio L, Martinotti G, Di Nicola M, Ruggeri F, Conte G, Di Giannantonio M, & Janiri L (2014). Targeting the glutamatergic system to treat pathological gambling: current evidence and future perspectives. BioMed Research International, 2014, 109786. 10.1155/2014/109786 [DOI] [PMC free article] [PubMed] [Google Scholar]
  345. Piszczek L, Constantinescu A, Kargl D, Lazovic J, Pekcec A, Nicholson JR, & Haubensak W (2022). Dissociation of impulsive traits by subthalamic metabotropic glutamate receptor 4. eLife, 11, e62123. 10.7554/eLife.62123 [DOI] [PMC free article] [PubMed] [Google Scholar]
  346. Pitts RC, & Febbo SM (2004). Quantitative analyses of methamphetamine’s effects on self-control choices: implications for elucidating behavioral mechanisms of drug action. Behavioural Processes, 66(3), 213–233. 10.1016/j.beproc.2004.03.006 [DOI] [PubMed] [Google Scholar]
  347. Popik P, Holuj M, Nikiforuk A, Kos T, Trullas R, & Skolnick P (2015). 1-aminocyclopropanecarboxylic acid (ACPC) produces procognitive but not antipsychotic-like effects in rats. Psychopharmacology, 232(6), 1025–1038. 10.1007/s00213-014-3738-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  348. Pothuizen HH, Jongen-Rêlo AL, Feldon J, & Yee BK (2005). Double dissociation of the effects of selective nucleus accumbens core and shell lesions on impulsive-choice behaviour and salience learning in rats. The European Journal of Neuroscience, 22(10), 2605–2616. 10.1111/j.1460-9568.2005.04388.x [DOI] [PubMed] [Google Scholar]
  349. Powers B, Joyce C, Kleinman JE, Hyde TM, Ajilore O, Leow A, & Sodhi MS (2020). Sex differences in the transcription of glutamate transporters in major depression and suicide. Journal of Affective Disorders, 277, 244–252. 10.1016/j.jad.2020.07.055 [DOI] [PubMed] [Google Scholar]
  350. Pozzi L, Baviera M, Sacchetti G, Calcagno E, Balducci C, Invernizzi RW, & Carli M (2011). Attention deficit induced by blockade of N-methyl D-aspartate receptors in the prefrontal cortex is associated with enhanced glutamate release and cAMP response element binding protein phosphorylation: role of metabotropic glutamate receptors 2/3. Neuroscience, 176, 336–348. 10.1016/j.neuroscience.2010.11.060 [DOI] [PubMed] [Google Scholar]
  351. Puts NA, Ryan M, Oeltzschner G, Horska A, Edden RAE, & Mahone EM (2020). Reduced striatal GABA in unmedicated children with ADHD at 7T. Psychiatry Research. Neuroimaging, 301, 111082. 10.1016/j.pscychresns.2020.111082 [DOI] [PubMed] [Google Scholar]
  352. Rachlin H, Raineri A, & Cross D (1991). Subjective probability and delay. The Journal of the Experimental Analysis of Behavior, 55(2), 233–244. 10.1901/jeab.1991.55-233 [DOI] [PMC free article] [PubMed] [Google Scholar]
  353. Radomsky ED, Haas GL, Mann JJ, & Sweeney JA (1999). Suicidal behavior in patients with schizophrenia and other psychotic disorders. The American Journal of Psychiatry, 156(10), 1590–1595. 10.1176/ajp.156.10.1590 [DOI] [PubMed] [Google Scholar]
  354. Rajala AZ, Jenison RL, & Populin LC (2015). Decision making: effects of methylphenidate on temporal discounting in nonhuman primates. Journal of Neurophysiology, 114(1), 70–79. 10.1152/jn.00278.2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  355. Rammes G, Rupprecht R, Ferrari U, Zieglgänsberger W, & Parsons CG (2001). The N-methyl-D-aspartate receptor channel blockers memantine, MRZ 2/579 and other amino-alkyl-cyclohexanes antagonise 5-HT(3) receptor currents in cultured HEK-293 and N1E-115 cell systems in a non-competitive manner. Neuroscience Letters, 306(1–2), 81–84. 10.1016/s0304-3940(01)01872-9 [DOI] [PubMed] [Google Scholar]
  356. Razoux F, Garcia R, & Léna I (2007). Ketamine, at a dose that disrupts motor behavior and latent inhibition, enhances prefrontal cortex synaptic efficacy and glutamate release in the nucleus accumbens. Neuropsychopharmacology, 32(3), 719–727. 10.1038/sj.npp.1301057 [DOI] [PubMed] [Google Scholar]
  357. Regan MC, Romero-Hernandez A, & Furukawa H (2015). A structural biology perspective on NMDA receptor pharmacology and function. Current Opinion in Structural Biology, 33, 68–75. 10.1016/j.sbi.2015.07.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  358. Rimmele TS, & Rosenberg PA (2016). GLT-1: the elusive presynaptic glutamate transporter. Neurochemistry International, 98, 19–28. 10.1016/j.neuint.2016.04.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  359. Ritter C, Buchmann A, Müller ST, Volleberg M, Haynes M, Ghisleni C, Noeske R, Tuura R, & Hasler G (2022). Evaluation of prefrontal γ-aminobutyric acid and glutamate levels in individuals with major depressive disorder using proton magnetic resonance spectroscopy. JAMA Psychiatry, 79(12), 1209–1216. 10.1001/jamapsychiatry.2022.3384 [DOI] [PMC free article] [PubMed] [Google Scholar]
  360. Rizzo F, Abaei A, Nespoli E, Fegert JM, Hengerer B, Rasche V, & Boeckers TM (2017). Aripiprazole and Riluzole treatment alters behavior and neurometabolites in young ADHD rats: a longitudinal 1H-NMR spectroscopy study at 11.7T. Translational Psychiatry, 7(8), e1189. 10.1038/tp.2017.167 [DOI] [PMC free article] [PubMed] [Google Scholar]
  361. Roberts H, Pozzi E, Vijayakumar N, Richmond S, Bray K, Deane C, & Whittle S (2021). Structural brain development and aggression: a longitudinal study in late childhood. Cognitive, Affective & Behavioral Neuroscience, 21(2), 401–411. 10.3758/s13415-021-00871-3 [DOI] [PubMed] [Google Scholar]
  362. Rocha GS, Freire MAM, Britto AM, Paiva KM, Oliveira RF, Fonseca IAT, Araújo DP, Oliveira LC, Guzen FP, Morais PLAG, & Cavalcanti JRLP (2023). Basal ganglia for beginners: the basic concepts you need to know and their role in movement control. Frontiers in Systems Neuroscience, 17, 1242929. 10.3389/fnsys.2023.1242929 [DOI] [PMC free article] [PubMed] [Google Scholar]
  363. Roland PE (1984). Organization of motor control by the normal human brain. Human Neurobiology, 2(4), 205–216. [PubMed] [Google Scholar]
  364. Rolls ET (2019). The cingulate cortex and limbic systems for emotion, action, and memory. Brain Structure & Function, 224(9), 3001–3018. 10.1007/s00429-019-01945-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  365. Rolls ET, Cheng W, & Feng J (2020). The orbitofrontal cortex: reward, emotion and depression. Brain Communications, 2(2), fcaa196. 10.1093/braincomms/fcaa196 [DOI] [PMC free article] [PubMed] [Google Scholar]
  366. Roozen HG, van der Kroft P, van Marle HJ, & Franken IH (2011). The impact of craving and impulsivity on aggression in detoxified cocaine-dependent patients. Journal of Substance Abuse Treatment, 40(4), 414–418. 10.1016/j.jsat.2010.12.003 [DOI] [PubMed] [Google Scholar]
  367. Rüsch N, Boeker M, Büchert M, Glauche V, Bohrmann C, Ebert D, Lieb K, Hennig J, & Tebartz Van Elst L (2010). Neurochemical alterations in women with borderline personality disorder and comorbid attention-deficit hyperactivity disorder. The World Journal of Biological Psychiatry, 11(2 Pt 2), 372–381. 10.3109/15622970801958331 [DOI] [PubMed] [Google Scholar]
  368. Russell VA (2001). Increased AMPA receptor function in slices containing the prefrontal cortex of spontaneously hypertensive rats. Metabolic Brain Disease, 16(3–4), 143–149. 10.1023/a:1012584826144 [DOI] [PubMed] [Google Scholar]
  369. Russell VA (2003). In vitro glutamate-stimulated release of dopamine from nucleus accumbens core and shell of spontaneously hypertensive rats. Metabolic Brain Disease, 18(2), 161–168. 10.1023/a:1023819220840 [DOI] [PubMed] [Google Scholar]
  370. Ryden MB, Bossenmaier M, & McLachlan C (1991). Aggressive behavior in cognitively impaired nursing home residents. Research in Nursing & Health, 14(2), 87–95. 10.1002/nur.4770140203 [DOI] [PubMed] [Google Scholar]
  371. Sable HJK, Lester DB, Potter JL, Nolen HG, Cruthird DM, Estes LM, Johnson AD, Regan SL, Williams MT, & Vorhees CV (2021). An assessment of executive function in two different rat models of attention-deficit hyperactivity disorder: Spontaneously hypertensive versus Lphn3 knockout rats. Genes, Brain, and Behavior, 20(8), e12767. 10.1111/gbb.12767 [DOI] [PMC free article] [PubMed] [Google Scholar]
  372. Sagvolden T (2000). Behavioral validation of the spontaneously hypertensive rat (SHR) as an animal model of attention-deficit/hyperactivity disorder (AD/HD). Neuroscience and Biobehavioral Reviews, 24(1), 31–39. 10.1016/s0149-7634(99)00058-5 [DOI] [PubMed] [Google Scholar]
  373. Sagvolden T, & Xu T (2008). l-Amphetamine improves poor sustained attention while d-amphetamine reduces overactivity and impulsiveness as well as improves sustained attention in an animal model of Attention-Deficit/Hyperactivity Disorder (ADHD). Behavioral and Brain Functions, 4, 3. 10.1186/1744-9081-4-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  374. Sakurai Y, & Sugimoto S (1985). Effects of lesions of prefrontal cortex and dorsomedial thalamus on delayed go/no-go alternation in rats. Behavioural Brain Research, 17(3), 213–219. 10.1016/0166-4328(85)90045-2 [DOI] [PubMed] [Google Scholar]
  375. Salgado S, & Kaplitt MG (2015). The nucleus accumbens: a comprehensive review. Stereotactic and Functional Neurosurgery, 93(2), 75–93. 10.1159/000368279 [DOI] [PubMed] [Google Scholar]
  376. Sanabria F, & Killeen PR (2008). Evidence for impulsivity in the Spontaneously Hypertensive Rat drawn from complementary response-withholding tasks. Behavioral and Brain Functions, 4, 7. 10.1186/1744-9081-4-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  377. Sarawagi A, Soni ND, & Patel AB (2021). Glutamate and GABA homeostasis and neurometabolism in major depressive disorder. Frontiers in Psychiatry, 12, 637863. 10.3389/fpsyt.2021.637863 [DOI] [PMC free article] [PubMed] [Google Scholar]
  378. Sari SA, Ulger D, Ersan S, Bakir D, Uzun Cicek A, & Ismailoglu F (2020). Effects of agmatine, glutamate, arginine, and nitric oxide on executive functions in children with attention deficit hyperactivity disorder. Journal of Neural Transmission, 127(12), 1675–1684. 10.1007/s00702-020-02261-4 [DOI] [PubMed] [Google Scholar]
  379. Saxena K, Howe M, Simeonova D, Steiner H, & Chang K (2006). Divalproex sodium reduces overall aggression in youth at high risk for bipolar disorder. Journal of Child and Adolescent Psychopharmacology, 16(3), 252–259. 10.1089/cap.2006.16.252 [DOI] [PubMed] [Google Scholar]
  380. Scarr E, Pavey G, Sundram S, MacKinnon A, & Dean B (2003). Decreased hippocampal NMDA, but not kainate or AMPA receptors in bipolar disorder. Bipolar Disorders, 5(4), 257–264. 10.1034/j.1399-5618.2003.00024.x [DOI] [PubMed] [Google Scholar]
  381. Schmaal L, Goudriaan AE, van der Meer J, van den Brink W, & Veltman DJ (2012a). The association between cingulate cortex glutamate concentration and delay discounting is mediated by resting state functional connectivity. Brain and Behavior, 2(5), 553–562. 10.1002/brb3.74 [DOI] [PMC free article] [PubMed] [Google Scholar]
  382. Schmaal L, van Harmelen AL, Chatzi V, Lippard ETC, Toenders YJ, Averill LA, Mazure CM, & Blumberg HP (2020). Imaging suicidal thoughts and behaviors: a comprehensive review of 2 decades of neuroimaging studies. Molecular Psychiatry, 25(2), 408–427. 10.1038/s41380-019-0587-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  383. Schmaal L, Veltman DJ, Nederveen A, van den Brink W, & Goudriaan AE (2012b). N-acetylcysteine normalizes glutamate levels in cocaine-dependent patients: a randomized crossover magnetic resonance spectroscopy study. Neuropsychopharmacology, 37(9), 2143–2152. 10.1038/npp.2012.66 [DOI] [PMC free article] [PubMed] [Google Scholar]
  384. Schousboe A, Scafidi S, Bak LK, Waagepetersen HS, & McKenna MC (2014). Glutamate metabolism in the brain focusing on astrocytes. Advances in Neurobiology, 11, 13–30. 10.1007/978-3-319-08894-5_2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  385. Schwartz TL, Sachdeva S, & Stahl SM (2012). Glutamate neurocircuitry: theoretical underpinnings in schizophrenia. Frontiers in Pharmacology, 3, 195. 10.3389/fphar.2012.00195 [DOI] [PMC free article] [PubMed] [Google Scholar]
  386. Schweizer N, Pupe S, Arvidsson E, Nordenankar K, Smith-Anttila CJ, Mahmoudi S, Andrén A, Dumas S, Rajagopalan A, Lévesque D, Leão RN, & Wallén-Mackenzie Å (2014). Limiting glutamate transmission in a Vglut2-expressing subpopulation of the subthalamic nucleus is sufficient to cause hyperlocomotion. Proceedings of the National Academy of Sciences of the United States of America, 111(21), 7837–7842. 10.1073/pnas.1323499111 [DOI] [PMC free article] [PubMed] [Google Scholar]
  387. Scofield MD, Heinsbroek JA, Gipson CD, Kupchik YM, Spencer S, Smith AC, Roberts-Wolfe D, & Kalivas PW (2016). The nucleus accumbens: mechanisms of addiction across drug classes reflect the importance of glutamate homeostasis. Pharmacological Reviews, 68(3), 816–871. 10.1124/pr.116.012484 [DOI] [PMC free article] [PubMed] [Google Scholar]
  388. Scott GA, Roebuck AJ, Greba Q, & Howland JG (2019). Performance of the trial-unique, delayed non-matching-to-location (TUNL) task depends on AMPA/Kainate, but not NMDA, ionotropic glutamate receptors in the rat posterior parietal cortex. Neurobiology of Learning and Memory, 159, 16–23. 10.1016/j.nlm.2019.02.001 [DOI] [PubMed] [Google Scholar]
  389. Selemon LD (2013). A role for synaptic plasticity in the adolescent development of executive function. Translational Psychiatry, 3(3), e238. 10.1038/tp.2013.7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  390. Semenova S, & Markou A (2007). The effects of the mGluR5 antagonist MPEP and the mGluR2/3 antagonist LY341495 on rats’ performance in the 5-choice serial reaction time task. Neuropharmacology, 52(3), 863–872. 10.1016/j.neuropharm.2006.10.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  391. Seo D, Patrick CJ, & Kennealy PJ (2008). Role of serotonin and dopamine system interactions in the neurobiology of impulsive aggression and its comorbidity with other clinical disorders. Aggression and Violent Behavior, 13(5), 383–395. 10.1016/j.avb.2008.06.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  392. Serafini G, Calcagno P, Lester D, Girardi P, Amore M, & Pompili M (2016). Suicide risk in Alzheimer’s disease: a systematic review. Current Alzheimer Research, 13(10), 1083–1099. 10.2174/1567205013666160720112608 [DOI] [PubMed] [Google Scholar]
  393. Sergerie K, Chochol C, & Armony JL (2008). The role of the amygdala in emotional processing: a quantitative meta-analysis of functional neuroimaging studies. Neuroscience and Biobehavioral Reviews, 32(4), 811–830. 10.1016/j.neubiorev.2007.12.002 [DOI] [PubMed] [Google Scholar]
  394. Sesia T, Temel Y, Lim LW, Blokland A, Steinbusch HW, & Visser-Vandewalle V (2008). Deep brain stimulation of the nucleus accumbens core and shell: opposite effects on impulsive action. Experimental Neurology, 214(1), 135–139. 10.1016/j.expneurol.2008.07.015 [DOI] [PubMed] [Google Scholar]
  395. Shan Y, Jia Y, Zhong S, Li X, Zhao H, Chen J, Lu Q, Zhang L, Li Z, Lai S, & Wang Y (2018). Correlations between working memory impairment and neurometabolites of prefrontal cortex and lenticular nucleus in patients with major depressive disorder. Journal of Affective Disorders, 227, 236–242. 10.1016/j.jad.2017.10.030 [DOI] [PubMed] [Google Scholar]
  396. Shepherd JD, & Huganir RL (2007). The cell biology of synaptic plasticity: AMPA receptor trafficking. Annual Review of Cell and Developmental Biology, 23, 613–643. 10.1146/annurev.cellbio.23.090506.123516 [DOI] [PubMed] [Google Scholar]
  397. Shin BC, Cepeda C, Eghbali M, Byun SY, Levine MS, & Devaskar SU (2021). Adult glut3 homozygous null mice survive to demonstrate neural excitability and altered neurobehavioral responses reminiscent of neurodevelopmental disorders. Experimental Neurology, 338, 113603. 10.1016/j.expneurol.2021.113603 [DOI] [PMC free article] [PubMed] [Google Scholar]
  398. Shiraishi-Yamaguchi Y, & Furuichi T (2007). The Homer family proteins. Genome Biology, 8(2), 206. 10.1186/gb-2007-8-2-206 [DOI] [PMC free article] [PubMed] [Google Scholar]
  399. Shirayama Y, Takahashi M, Osone F, Hara A, & Okubo T (2017). Myo-inositol, glutamate, and glutamine in the prefrontal cortex, hippocampus, and amygdala in major depression. Biological Psychiatry. Cognitive Neuroscience and Neuroimaging, 2(2), 196–204. 10.1016/j.bpsc.2016.11.006 [DOI] [PubMed] [Google Scholar]
  400. Siddiqui SV, Chatterjee U, Kumar D, Siddiqui A, & Goyal N (2008). Neuropsychology of prefrontal cortex. Indian Journal of Psychiatry, 50(3), 202–208. 10.4103/0019-5545.43634 [DOI] [PMC free article] [PubMed] [Google Scholar]
  401. Simon NW, Beas BS, Montgomery KS, Haberman RP, Bizon JL, & Setlow B (2013). Prefrontal cortical-striatal dopamine receptor mRNA expression predicts distinct forms of impulsivity. The European Journal of Neuroscience, 37(11), 1779–1788. 10.1111/ejn.12191 [DOI] [PMC free article] [PubMed] [Google Scholar]
  402. Skalny AV, Mazaletskaya AL, Zaitseva IP, Skalny AA, Spandidos DA, Tsatsakis A, Lobanova YN, Skalnaya MG, Aschner M, & Tinkov AA (2021). Alterations in serum amino acid profiles in children with attention deficit/hyperactivity disorder. Biomedical Reports, 14(5), 47. 10.3892/br.2021.1423 [DOI] [PMC free article] [PubMed] [Google Scholar]
  403. Slotnick SD (2022). Does working memory activate the hippocampus during the late delay period?. Cognitive Neuroscience, 13(3–4), 182–207. 10.1080/17588928.2022.2075842 [DOI] [PubMed] [Google Scholar]
  404. Smaga I, Frankowska M, & Filip M (2021). N-acetylcysteine as a new prominent approach for treating psychiatric disorders. British Journal of Pharmacology, 178(13), 2569–2594. 10.1111/bph.15456 [DOI] [PubMed] [Google Scholar]
  405. Smaragdi A, Chavez S, Lobaugh NJ, Meyer JH, & Kolla NJ (2019). Differential levels of prefrontal cortex glutamate+glutamine in adults with antisocial personality disorder and bipolar disorder: a proton magnetic resonance spectroscopy study. Progress in Neuro-Psychopharmacology & Biological Psychiatry, 93, 250–255. 10.1016/j.pnpbp.2019.04.002 [DOI] [PubMed] [Google Scholar]
  406. Smesny S, Große J, Gussew A, Langbein K, Schönfeld N, Wagner G, Valente M, & Reichenbach JR (2018). Prefrontal glutamatergic emotion regulation is disturbed in cluster B and C personality disorders - a combined 1H/31P-MR spectroscopic study. Journal of Affective Disorders, 227, 688–697. 10.1016/j.jad.2017.10.044 [DOI] [PubMed] [Google Scholar]
  407. Smith KM (2018). Hyperactivity in mice lacking one allele of the glutamic acid decarboxylase 67 gene. Attention Deficit and Hyperactivity Disorders, 10(4), 267–271. 10.1007/s12402-018-0254-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  408. Smith BL, Laaker CJ, Lloyd KR, Hiltz AR, & Reyes TM (2020). Adolescent microglia play a role in executive function in male mice exposed to perinatal high fat diet. Brain, Behavior, and Immunity, 84, 80–89. 10.1016/j.bbi.2019.11.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  409. Soeiro-de-Souza MG, Henning A, Machado-Vieira R, Moreno RA, Pastorello BF, da Costa Leite C, Vallada H, & Otaduy MC (2015). Anterior cingulate Glutamate-Glutamine cycle metabolites are altered in euthymic bipolar I disorder. European Neuropsychopharmacology, 25(12), 2221–2229. 10.1016/j.euroneuro.2015.09.020 [DOI] [PubMed] [Google Scholar]
  410. Soeiro-de-Souza MG, Otaduy MCG, Machado-Vieira R, Moreno RA, Nery FG, Leite C, & Lafer B (2018). Anterior cingulate cortex glutamatergic metabolites and mood stabilizers in euthymic bipolar i disorder patients: a proton magnetic resonance spectroscopy study. Biological Psychiatry. Cognitive Neuroscience and Neuroimaging, 3(12), 985–991. 10.1016/j.bpsc.2018.02.007 [DOI] [PubMed] [Google Scholar]
  411. Soiza-Reilly M, & Commons KG (2011). Glutamatergic drive of the dorsal raphe nucleus. Journal of Chemical Neuroanatomy, 41(4), 247–255. 10.1016/j.jchemneu.2011.04.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  412. Soliva JC, Moreno A, Fauquet J, Bielsa A, Carmona S, Gispert JD, Rovira M, Bulbena A, & Vilarroya O (2010). Cerebellar neurometabolite abnormalities in pediatric attention/deficit hyperactivity disorder: a proton MR spectroscopic study. Neuroscience Letters, 470(1), 60–64. 10.1016/j.neulet.2009.12.056 [DOI] [PubMed] [Google Scholar]
  413. Somkuwar SS, Kantak KM, Bardo MT, & Dwoskin LP (2016). Adolescent methylphenidate treatment differentially alters adult impulsivity and hyperactivity in the Spontaneously Hypertensive Rat model of ADHD. Pharmacology, Biochemistry, and Behavior, 141, 66–77. 10.1016/j.pbb.2015.12.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  414. Sontag TA, Tucha O, Walitza S, & Lange KW (2010). Animal models of attention deficit/hyperactivity disorder (ADHD): a critical review. Attention Deficit and Hyperactivity Disorders, 2(1), 1–20. 10.1007/s12402-010-0019-x [DOI] [PubMed] [Google Scholar]
  415. Spieser L, van den Wildenberg W, Hasbroucq T, Ridderinkhof KR, & Burle B (2015). Controlling your impulses: electrical stimulation of the human supplementary motor complex prevents impulsive errors. The Journal of Neuroscience, 35(7), 3010–3015. 10.1523/JNEUROSCI.1642-14.2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  416. Squire LR, Genzel L, Wixted JT, & Morris RG (2015). Memory consolidation. Cold Spring Harbor Perspectives in Biology, 7(8), a021766. 10.1101/cshperspect.a021766 [DOI] [PMC free article] [PubMed] [Google Scholar]
  417. Stafstrom CE (2010). Mechanisms of action of antiepileptic drugs: the search for synergy. Current Opinion in Neurology, 23(2), 157–163. 10.1097/WCO.0b013e32833735b5 [DOI] [PubMed] [Google Scholar]
  418. Stark I, Rai D, Lundberg M, Culpin I, Nordström SI, Ohlis A, & Magnusson C (2022). Autism and self-harm: a population-based and discordant sibling study of young individuals. Acta Psychiatrica Scandinavica, 146(5), 468–477. 10.1111/acps.13479 [DOI] [PMC free article] [PubMed] [Google Scholar]
  419. Steiner J, Walter M, Gos T, Guillemin GJ, Bernstein HG, Sarnyai Z, Mawrin C, Brisch R, Bielau H, Meyer zu Schwabedissen L, Bogerts B, & Myint AM (2011). Severe depression is associated with increased microglial quinolinic acid in subregions of the anterior cingulate gyrus: evidence for an immune-modulated glutamatergic neurotransmission?. Journal of Neuroinflammation, 8, 94. 10.1186/1742-2094-8-94 [DOI] [PMC free article] [PubMed] [Google Scholar]
  420. Sterley TL, Howells FM, Dimatelis JJ, & Russell VA (2016). Genetic predisposition and early life experience interact to determine glutamate transporter (GLT1) and solute carrier family 12 member 5 (KCC2) levels in rat hippocampus. Metabolic Brain Disease, 31(1), 169–182. 10.1007/s11011-015-9742-5 [DOI] [PubMed] [Google Scholar]
  421. Stifani N (2014). Motor neurons and the generation of spinal motor neuron diversity. Frontiers in Cellular Neuroscience, 8, 293. 10.3389/fncel.2014.00293 [DOI] [PMC free article] [PubMed] [Google Scholar]
  422. Stojek MM, Fischer S, Murphy CM, & MacKillop J (2014). The role of impulsivity traits and delayed reward discounting in dysregulated eating and drinking among heavy drinkers. Appetite, 80, 81–88. 10.1016/j.appet.2014.05.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  423. Stopper CM, Green EB, & Floresco SB (2014). Selective involvement by the medial orbitofrontal cortex in biasing risky, but not impulsive, choice. Cerebral Cortex, 24(1), 154–162. 10.1093/cercor/bhs297 [DOI] [PubMed] [Google Scholar]
  424. Strickland JC, & Johnson MW (2021). Rejecting impulsivity as a psychological construct: A theoretical, empirical, and sociocultural argument. Psychological Review, 128(2), 336–361. 10.1037/rev0000263 [DOI] [PMC free article] [PubMed] [Google Scholar]
  425. Subramaniam P, Prescot A, McGlade E, Renshaw P, & Yurgelun-Todd D (2022). Examination of gamma-aminobutyric acid and glutamate-glutamine levels in association with impulsive behavior in adolescent marijuana users. Drug and Alcohol Dependence, 233, 109326. 10.1016/j.drugalcdep.2022.109326 [DOI] [PMC free article] [PubMed] [Google Scholar]
  426. Sukhotina IA, Dravolina OA, Novitskaya Y, Zvartau EE, Danysz W, & Bespalov AY (2008). Effects of mGlu1 receptor blockade on working memory, time estimation, and impulsivity in rats. Psychopharmacology, 196(2), 211–220. 10.1007/s00213-007-0953-2 [DOI] [PubMed] [Google Scholar]
  427. Surman CB, Hammerness PG, Petty C, Spencer T, Doyle R, Napolean S, Chu N, Yorks D, & Biederman J (2013). A pilot open label prospective study of memantine monotherapy in adults with ADHD. The World Journal of Biological Psychiatry, 14(4), 291–298. 10.3109/15622975.2011.623716 [DOI] [PubMed] [Google Scholar]
  428. Swann AC, Moeller FG, Steinberg JL, Schneider L, Barratt ES, & Dougherty DM (2007). Manic symptoms and impulsivity during bipolar depressive episodes. Bipolar Disorders, 9(3), 206–212. 10.1111/j.1399-5618.2007.00357.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  429. Swann AC, Steinberg JL, Lijffijt M, & Moeller FG (2008). Impulsivity: differential relationship to depression and mania in bipolar disorder. Journal of Affective Disorders, 106(3), 241–248. 10.1016/j.jad.2007.07.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  430. Szuhany KL, MacKenzie D Jr, & Otto MW (2018). The impact of depressed mood, working memory capacity, and priming on delay discounting. Journal of Behavior Therapy and Experimental Psychiatry, 60, 37–41. 10.1016/j.jbtep.2018.03.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  431. Takahashi T, Oono H, Inoue T, Boku S, Kako Y, Kitaichi Y, Kusumi I, Masui T, Nakagawa S, Suzuki K, Tanaka T, Koyama T, & Radford MH (2008). Depressive patients are more impulsive and inconsistent in intertemporal choice behavior for monetary gain and loss than healthy subjects--an analysis based on Tsallis’ statistics. Neuro Endocrinology Letters, 29(3), 351–358. [PubMed] [Google Scholar]
  432. Tanabe Y, Masu M, Ishii T, Shigemoto R, & Nakanishi S (1992). A family of metabotropic glutamate receptors. Neuron, 8(1), 169–179. 10.1016/0896-6273(92)90118-w [DOI] [PubMed] [Google Scholar]
  433. Tanno T, Maguire DR, Henson C, & France CP (2014). Effects of amphetamine and methylphenidate on delay discounting in rats: interactions with order of delay presentation. Psychopharmacology, 231(1), 85–95. 10.1007/s00213-013-3209-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  434. Terry AV Jr, Buccafusco JJ, Schade RF, Vandenhuerk L, Callahan PM, Beck WD, Hutchings EJ, Chapman JM, Li P, & Bartlett MG (2012). The nicotine metabolite, cotinine, attenuates glutamate (NMDA) antagonist-related effects on the performance of the five choice serial reaction time task (5C-SRTT) in rats. Biochemical Pharmacology, 83(7), 941–951. 10.1016/j.bcp.2011.12.043 [DOI] [PMC free article] [PubMed] [Google Scholar]
  435. Testa CM, Standaert DG, Young AB, & Penney JB Jr (1994). Metabotropic glutamate receptor mRNA expression in the basal ganglia of the rat. The Journal of Neuroscience, 14(5 Pt 2), 3005–3018. 10.1523/JNEUROSCI.14-05-03005.1994 [DOI] [PMC free article] [PubMed] [Google Scholar]
  436. Thoma MV, Maercker A, & Forstmeier S (2017). Evidence for different trajectories of delay discounting in older adults with mild cognitive impairment and mild Alzheimer’s disease. The Journals of Gerontology. Series B, Psychological Sciences and Social Sciences, 72(6), 956–965. 10.1093/geronb/gbw010 [DOI] [PubMed] [Google Scholar]
  437. Thomson PA, Duff B, Blackwood DH, Romaniuk L, Watson A, Whalley HC, Li X, Dauvermann MR, Moorhead TW, Bois C, Ryan NM, Redpath H, Hall L, Morris SW, van Beek EJ, Roberts N, Porteous DJ, St Clair D, Whitcher B, Dunlop J, … Lawrie SM (2016). Balanced translocation linked to psychiatric disorder, glutamate, and cortical structure/function. NPJ Schizophrenia, 2, 16024. 10.1038/npjschz.2016.24 [DOI] [PMC free article] [PubMed] [Google Scholar]
  438. Thompson M, Weickert CS, Wyatt E, & Webster MJ (2009). Decreased glutamic acid decarboxylase(67) mRNA expression in multiple brain areas of patients with schizophrenia and mood disorders. Journal of Psychiatric Research, 43(11), 970–977. 10.1016/j.jpsychires.2009.02.005 [DOI] [PubMed] [Google Scholar]
  439. Togashi H, Sakisaka T, & Takai Y (2009). Cell adhesion molecules in the central nervous system. Cell Adhesion & Migration, 3(1), 29–35. 10.4161/cam.3.1.6773 [DOI] [PMC free article] [PubMed] [Google Scholar]
  440. Torgner I, & Kvamme E (1990). Synthesis of transmitter glutamate and the glial-neuron interrelationship. Molecular and Chemical Neuropathology, 12(1), 11–17. 10.1007/BF03160053 [DOI] [PubMed] [Google Scholar]
  441. Toro C, & Deakin JF (2005). NMDA receptor subunit NRI and postsynaptic protein PSD-95 in hippocampus and orbitofrontal cortex in schizophrenia and mood disorder. Schizophrenia Research, 80(2–3), 323–330. 10.1016/j.schres.2005.07.003 [DOI] [PubMed] [Google Scholar]
  442. Tritt K, Nickel C, Lahmann C, Leiberich PK, Rother WK, Loew TH, & Nickel MK (2005). Lamotrigine treatment of aggression in female borderline-patients: a randomized, double-blind, placebo-controlled study. Journal of Psychopharmacology, 19(3), 287–291. 10.1177/0269881105051540 [DOI] [PubMed] [Google Scholar]
  443. Tsai GE, Ragan P, Chang R, Chen S, Linnoila VM, & Coyle JT (1998). Increased glutamatergic neurotransmission and oxidative stress after alcohol withdrawal. The American Journal of Psychiatry, 155(6), 726–732. 10.1176/ajp.155.6.726 [DOI] [PubMed] [Google Scholar]
  444. Tsutsui-Kimura I, Ohmura Y, Izumi T, Yamaguchi T, Yoshida T, & Yoshioka M (2009). The effects of serotonin and/or noradrenaline reuptake inhibitors on impulsive-like action assessed by the three-choice serial reaction time task: a simple and valid model of impulsive action using rats. Behavioural Pharmacology, 20(5–6), 474–483. 10.1097/FBP.0b013e3283305e65 [DOI] [PubMed] [Google Scholar]
  445. Turic D, Langley K, Mills S, Stephens M, Lawson D, Govan C, Williams N, Van Den Bree M, Craddock N, Kent L, Owen M, O’Donovan M, & Thapar A (2004). Follow-up of genetic linkage findings on chromosome 16p13: evidence of association of N-methyl-D aspartate glutamate receptor 2A gene polymorphism with ADHD. Molecular Psychiatry, 9(2), 169–173. 10.1038/sj.mp.4001387 [DOI] [PubMed] [Google Scholar]
  446. Ucha M, Roura-Martínez D, Contreras A, Pinto-Rivero S, Orihuel J, Ambrosio E, & Higuera-Matas A (2019). Impulsive action and impulsive choice are differentially associated with gene expression variations of the GABAA receptor alfa 1 subunit and the CB1 receptor in the lateral and medial orbitofrontal cortices. Frontiers in Behavioral Neuroscience, 13, 22. 10.3389/fnbeh.2019.00022 [DOI] [PMC free article] [PubMed] [Google Scholar]
  447. Uezato A, Meador-Woodruff JH, & McCullumsmith RE (2009). Vesicular glutamate transporter mRNA expression in the medial temporal lobe in major depressive disorder, bipolar disorder, and schizophrenia. Bipolar Disorders, 11(7), 711–725. 10.1111/j.1399-5618.2009.00752.x [DOI] [PubMed] [Google Scholar]
  448. Umehara H, Numata S, Watanabe SY, Hatakeyama Y, Kinoshita M, Tomioka Y, Nakahara K, Nikawa T, & Ohmori T (2017). Altered KYN/TRP, Gln/Glu, and Met/methionine sulfoxide ratios in the blood plasma of medication-free patients with major depressive disorder. Scientific Reports, 7(1), 4855. 10.1038/s41598-017-05121-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  449. Utter AA, & Basso MA (2008). The basal ganglia: an overview of circuits and function. Neuroscience and Biobehavioral Reviews, 32(3), 333–342. 10.1016/j.neubiorev.2006.11.003 [DOI] [PubMed] [Google Scholar]
  450. van Amen-Hellebrekers CJ, Jansen S, Pfundt R, Schuurs-Hoeijmakers JH, Koolen DA, Marcelis CL, de Leeuw N, & de Vries BB (2016). Duplications of SLC1A3: associated with ADHD and autism. European Journal of Medical Genetics, 59(8), 373–376. 10.1016/j.ejmg.2016.06.003 [DOI] [PubMed] [Google Scholar]
  451. Vandenbeuch A, & Kinnamon SC (2016). Glutamate: tastant and neuromodulator in taste buds. Advances in Nutrition, 7(4), 823S–7S. 10.3945/an.115.011304 [DOI] [PMC free article] [PubMed] [Google Scholar]
  452. van der Veen B, Kapanaiah SKT, Kilonzo K, Steele-Perkins P, Jendryka MM, Schulz S, Tasic B, Yao Z, Zeng H, Akam T, Nicholson JR, Liss B, Nissen W, Pekcec A, & Kätzel D (2021). Control of impulsivity by Gi-protein signalling in layer-5 pyramidal neurons of the anterior cingulate cortex. Communications Biology, 4(1), 662. 10.1038/s42003-021-02188-w [DOI] [PMC free article] [PubMed] [Google Scholar]
  453. Van Dessel J, Sonuga-Barke E, Mies G, Lemiere J, Van der Oord S, Morsink S, & Danckaerts M (2018). Delay aversion in attention deficit/hyperactivity disorder is mediated by amygdala and prefrontal cortex hyper-activation. Journal of Child Psychology and Psychiatry, 59(8), 888–899. 10.1111/jcpp.12868 [DOI] [PubMed] [Google Scholar]
  454. van Heukelum S, Mogavero F, van de Wal MAE, Geers FE, França ASC, Buitelaar JK, Beckmann CF, Glennon JC, & Havenith MN (2019). Gradient of parvalbumin- and somatostatin-expressing interneurons across cingulate cortex is differentially linked to aggression and sociability in BALB/cJ mice. Frontiers in Psychiatry, 10, 809. 10.3389/fpsyt.2019.00809 [DOI] [PMC free article] [PubMed] [Google Scholar]
  455. van Heukelum S, Tulva K, Geers FE, van Dulm S, Ruisch IH, Mill J, Viana JF, Beckmann CF, Buitelaar JK, Poelmans G, Glennon JC, Vogt BA, Havenith MN, & França ASC (2021). A central role for anterior cingulate cortex in the control of pathological aggression. Current Biology, 31(11), 2321–2333.e5. 10.1016/j.cub.2021.03.062 [DOI] [PubMed] [Google Scholar]
  456. Varea E, Guirado R, Gilabert-Juan J, Martí U, Castillo-Gomez E, Blasco-Ibáñez JM, Crespo C, & Nacher J (2012). Expression of PSA-NCAM and synaptic proteins in the amygdala of psychiatric disorder patients. Journal of Psychiatric Research, 46(2), 189–197. 10.1016/j.jpsychires.2011.10.011 [DOI] [PubMed] [Google Scholar]
  457. Verbruggen F, & McLaren R (2018). Effects of reward and punishment on the interaction between going and stopping in a selective stop-change task. Psychological Research, 82(2), 353–370. 10.1007/s00426-016-0827-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  458. Vieira M, Yong XLH, Roche KW, & Anggono V (2020). Regulation of NMDA glutamate receptor functions by the GluN2 subunits. Journal of Neurochemistry, 154(2), 121–143. 10.1111/jnc.14970 [DOI] [PMC free article] [PubMed] [Google Scholar]
  459. Viggiano D, Vallone D, Welzl H, & Sadile AG (2002). The Naples High- and Low-Excitability rats: selective breeding, behavioral profile, morphometry, and molecular biology of the mesocortical dopamine system. Behavior Genetics, 32(5), 315–333. 10.1023/a:1020210221156 [DOI] [PubMed] [Google Scholar]
  460. Voon V, Chang-Webb YC, Morris LS, Cooper E, Sethi A, Baek K, Grant J, Robbins TW, & Harrison NA (2015). Waiting impulsivity: the influence of acute methylphenidate and feedback. The International Journal of Neuropsychopharmacology, 19(1), pyv074. 10.1093/ijnp/pyv074 [DOI] [PMC free article] [PubMed] [Google Scholar]
  461. Vos M, & Hartman CA (2022). The decreasing prevalence of ADHD across the adult lifespan confirmed. Journal of Global Health, 12, 03024. 10.7189/jogh.12.03024 [DOI] [PMC free article] [PubMed] [Google Scholar]
  462. Wang YG, Chen S, Xu ZM, Shen ZH, Wang YQ, He XY, Cao RF, Roberts DL, Shi JF, & Wang YQ (2017a). Family history of suicide and high motor impulsivity distinguish suicide attempters from suicide ideators among college students. Journal of Psychiatric Research, 90, 21–25. 10.1016/j.jpsychires.2017.02.006 [DOI] [PubMed] [Google Scholar]
  463. Wang LJ, Lin LC, Lee SY, Wu CC, Chou WJ, Hsu CF, Tseng HH, & Lin WC (2021). l-Cystine is associated with the dysconnectivity of the default-mode network and salience network in attention-deficit/hyperactivity disorder. Psychoneuroendocrinology, 125, 105105. 10.1016/j.psyneuen.2020.105105 [DOI] [PubMed] [Google Scholar]
  464. Wang H, Lesh TA, Maddock RJ, Fassbender C, & Carter CS (2020a). Delay discounting abnormalities are seen in first-episode schizophrenia but not in bipolar disorder. Schizophrenia Research, 216, 200–206. 10.1016/j.schres.2019.11.063 [DOI] [PMC free article] [PubMed] [Google Scholar]
  465. Wang R, & Reddy PH (2017). Role of glutamate and NMDA receptors in Alzheimer’s disease. Journal of Alzheimer’s Disease, 57(4), 1041–1048. 10.3233/JAD-160763 [DOI] [PMC free article] [PubMed] [Google Scholar]
  466. Warnell KR, Maniscalco S, Baker S, Yi R, & Redcay E (2019). Social and delay discounting in autism spectrum disorder. Autism Research, 12(6), 870–877. 10.1002/aur.2085 [DOI] [PMC free article] [PubMed] [Google Scholar]
  467. Warr O, Takahashi M, & Attwell D (1999). Modulation of extracellular glutamate concentration in rat brain slices by cystine-glutamate exchange. The Journal of Physiology, 514 (Pt 3)(Pt 3), 783–793. 10.1111/j.1469-7793.1999.783ad.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  468. Warton FL, Howells FM, & Russell VA (2009). Increased glutamate-stimulated release of dopamine in substantia nigra of a rat model for attention-deficit/hyperactivity disorder--lack of effect of methylphenidate. Metabolic Brain Disease, 24(4), 599–613. 10.1007/s11011-009-9166-1 [DOI] [PubMed] [Google Scholar]
  469. Weafer J, Dzemidzic M, Eiler W 2nd, Oberlin BG, Wang Y, & Kareken DA (2015). Associations between regional brain physiology and trait impulsivity, motor inhibition, and impaired control over drinking. Psychiatry Research, 233(2), 81–87. 10.1016/j.pscychresns.2015.04.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  470. Wei ZX, Wu Q, Liu QS, & Cheng Y (2020). Neurotransmitter system aberrations in patients with drug addiction. Journal of Neural Transmission, 127(12), 1641–1650. 10.1007/s00702-020-02242-7 [DOI] [PubMed] [Google Scholar]
  471. Weisz CJC, Williams SG, Eckard CS, Divito CB, Ferreira DW, Fantetti KN, Dettwyler SA, Cai HM, Rubio ME, Kandler K, & Seal RP (2021). Outer hair cell glutamate signaling through type ii spiral ganglion afferents activates neurons in the cochlear nucleus in response to nondamaging sounds. The Journal of Neuroscience, 41(13), 2930–2943. 10.1523/JNEUROSCI.0619-20.2021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  472. Wenzel JM, Zlebnik NE, Patton MH, Smethells JR, Ayvazian VM, Dantrassy HM, Zhang LY, Mathur BN, & Cheer JF (2023). Selective chemogenetic inactivation of corticoaccumbal projections disrupts trait choice impulsivity. Neuropsychopharmacology, 48(12), 1821–1831. 10.1038/s41386-023-01604-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  473. Whiteside SP, & Lynam DR (2001). The Five Factor Model and impulsivity: Using a structural model of personality to understand impulsivity. Personality and Individual Differences, 30(4), 669–689. 10.1016/S0191-8869(00)00064-7 [DOI] [Google Scholar]
  474. Wikenheiser AM, & Schoenbaum G (2016). Over the river, through the woods: cognitive maps in the hippocampus and orbitofrontal cortex. Nature Reviews. Neuroscience, 17(8), 513–523. 10.1038/nrn.2016.56 [DOI] [PMC free article] [PubMed] [Google Scholar]
  475. Wilbertz G, van Elst LT, Delgado MR, Maier S, Feige B, Philipsen A, & Blechert J (2012). Orbitofrontal reward sensitivity and impulsivity in adult attention deficit hyperactivity disorder. NeuroImage, 60(1), 353–361. 10.1016/j.neuroimage.2011.12.011 [DOI] [PubMed] [Google Scholar]
  476. Winstanley CA (2011). The utility of rat models of impulsivity in developing pharmacotherapies for impulse control disorders. British Journal of Pharmacology, 164(4), 1301–1321. 10.1111/j.1476-5381.2011.01323.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  477. Winstanley CA, Theobald DE, Cardinal RN, & Robbins TW (2004). Contrasting roles of basolateral amygdala and orbitofrontal cortex in impulsive choice. The Journal of Neuroscience, 24(20), 4718–4722. 10.1523/JNEUROSCI.5606-03.2004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  478. Wischhof L, Hollensteiner KJ, & Koch M (2011). Impulsive behaviour in rats induced by intracortical DOI infusions is antagonized by co-administration of an mGlu2/3 receptor agonist. Behavioural Pharmacology, 22(8), 805–813. 10.1097/FBP.0b013e32834d6279 [DOI] [PubMed] [Google Scholar]
  479. Wischhof L, & Koch M (2012). Pre-treatment with the mGlu2/3 receptor agonist LY379268 attenuates DOI-induced impulsive responding and regional c-Fos protein expression. Psychopharmacology, 219(2), 387–400. 10.1007/s00213-011-2441-y [DOI] [PubMed] [Google Scholar]
  480. Wittmann M, Lovero KL, Lane SD, & Paulus MP (2010). Now or later? Striatum and insula activation to immediate versus delayed rewards. Journal of Neuroscience, Psychology, and Economics, 3(1), 15–26. 10.1037/a0017252 [DOI] [PMC free article] [PubMed] [Google Scholar]
  481. Woolverton WL, Myerson J, & Green L (2007). Delay discounting of cocaine by rhesus monkeys. Experimental and Clinical Psychopharmacology, 15(3), 238–244. 10.1037/1064-1297.15.3.238 [DOI] [PMC free article] [PubMed] [Google Scholar]
  482. Wrangham RW (2018). Two types of aggression in human evolution. Proceedings of the National Academy of Sciences of the United States of America, 115(2), 245–253. 10.1073/pnas.1713611115 [DOI] [PMC free article] [PubMed] [Google Scholar]
  483. Wu S, Yin Y, & Du L (2022). Blood-brain barrier dysfunction in the pathogenesis of major depressive disorder. Cellular and Molecular Neurobiology, 42(8), 2571–2591. 10.1007/s10571-021-01153-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  484. Yabuki Y, Shioda N, Maeda T, Hiraide S, Togashi H, & Fukunaga K (2014). Aberrant CaMKII activity in the medial prefrontal cortex is associated with cognitive dysfunction in ADHD model rats. Brain Research, 1557, 90–100. 10.1016/j.brainres.2014.02.025 [DOI] [PubMed] [Google Scholar]
  485. Yael D, Tahary O, Gurovich B, Belelovsky K, & Bar-Gad I (2019). Disinhibition of the nucleus accumbens leads to macro-scale hyperactivity consisting of micro-scale behavioral segments encoded by striatal activity. The Journal of Neuroscience, 39(30), 5897–5909. 10.1523/JNEUROSCI.3120-18.2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  486. Yan WS, Zhang RR, Lan Y, Li YH, & Sui N (2016). Comparison of impulsivity in non-problem, at-risk and problem gamblers. Scientific Reports, 6, 39233. 10.1038/srep39233 [DOI] [PMC free article] [PubMed] [Google Scholar]
  487. Yan WS, Zheng DH, & Liu MM (2022). trait impulsivity and choice impulsivity in young adult students with probable binge eating disorder. Frontiers in Psychiatry, 13, 838700. 10.3389/fpsyt.2022.838700 [DOI] [PMC free article] [PubMed] [Google Scholar]
  488. Yates JR (2018). Dissecting drug effects in preclinical models of impulsive choice: emphasis on glutamatergic compounds. Psychopharmacology, 235(3), 607–626. 10.1007/s00213-017-4825-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  489. Yates JR, & Bardo MT (2017). Effects of intra-accumbal administration of dopamine and ionotropic glutamate receptor drugs on delay discounting performance in rats. Behavioral Neuroscience, 131(5), 392–405. 10.1037/bne0000214 [DOI] [PMC free article] [PubMed] [Google Scholar]
  490. Yates JR, Batten SR, Bardo MT, & Beckmann JS (2015). Role of ionotropic glutamate receptors in delay and probability discounting in the rat. Psychopharmacology, 232(7), 1187–1196. 10.1007/s00213-014-3747-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  491. Yates JR, Day HA, Evans KE, Igwe HO, Kappesser JL, Miller AL, Murray CP, Torline BT, Ellis AL, & Stacy WL (2019). Effects of d-amphetamine and MK-801 on impulsive choice: modulation by schedule of reinforcement and delay length. Behavioural Brain Research, 376, 112228. 10.1016/j.bbr.2019.112228 [DOI] [PMC free article] [PubMed] [Google Scholar]
  492. Yates JR, Gunkel BT, Rogers KK, Hughes MN, & Prior NA (2017a). Effects of N-methyl-D-aspartate receptor ligands on sensitivity to reinforcer magnitude and delayed reinforcement in a delay-discounting procedure. Psychopharmacology, 234(3), 461–473. 10.1007/s00213-016-4469-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  493. Yates JR, Prior NA, Chitwood MR, Day HA, Heidel JR, Hopkins SE, Muncie BT, Paradella-Bradley TA, Sestito AP, Vecchiola AN, & Wells EE (2018). Effects of GluN2B-selective antagonists on delay and probability discounting in male rats: modulation by delay/probability presentation order. Experimental and Clinical Psychopharmacology, 26(6), 525–540. 10.1037/pha0000216 [DOI] [PMC free article] [PubMed] [Google Scholar]
  494. Yates JR, Rogers KK, Gunkel BT, Prior NA, Hughes MN, Sharpe SM, Campbell HL, Johnson AB, Keller MG, Breitenstein KA, & Shults HN (2017b). Effects of Group I metabotropic glutamate receptor antagonists on sensitivity to reinforcer magnitude and delayed reinforcement in a delay-discounting task in rats: Contribution of delay presentation order. Behavioural Brain Research, 322(Pt A), 29–33. 10.1016/j.bbr.2017.01.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  495. Yin HH, & Knowlton BJ (2006). The role of the basal ganglia in habit formation. Nature Reviews. Neuroscience, 7(6), 464–476. 10.1038/nrn1919 [DOI] [PubMed] [Google Scholar]
  496. Yoon T, Okada J, Jung MW, & Kim JJ (2008). Prefrontal cortex and hippocampus subserve different components of working memory in rats. Learning & Memory, 15(3), 97–105. 10.1101/lm.850808 [DOI] [PMC free article] [PubMed] [Google Scholar]
  497. Yu LQ, Lee S, Katchmar N, Satterthwaite TD, Kable JW, & Wolf DH (2017). Steeper discounting of delayed rewards in schizophrenia but not first-degree relatives. Psychiatry Research, 252, 303–309. 10.1016/j.psychres.2017.02.062 [DOI] [PMC free article] [PubMed] [Google Scholar]
  498. Zald DH, & Kim SW (1996). Anatomy and function of the orbital frontal cortex, I: anatomy, neurocircuitry; and obsessive-compulsive disorder. The Journal of Neuropsychiatry and Clinical Neurosciences, 8(2), 125–138. 10.1176/jnp.8.2.125 [DOI] [PubMed] [Google Scholar]
  499. Zanatta G, Sula A, Miles AJ, Ng LCT, Torella R, Pryde DC, DeCaen PG, & Wallace BA (2019). Valproic acid interactions with the NavMs voltage-gated sodium channel. Proceedings of the National Academy of Sciences of the United States of America, 116(52), 26549–26554. 10.1073/pnas.1909696116 [DOI] [PMC free article] [PubMed] [Google Scholar]
  500. Zaremba D, Schulze Kalthoff I, Förster K, Redlich R, Grotegerd D, Leehr EJ, Meinert S, Dohm K, Bürger C, Enneking V, Böhnlein J, Repple J, Opel N, Jörgens S, Yüksel D, Schmitt S, Stein F, Kircher T, Krug A, Nenadić I, … Dannlowski U (2019). The effects of processing speed on memory impairment in patients with major depressive disorder. Progress in Neuro-Psychopharmacology & Biological Psychiatry, 92, 494–500. 10.1016/j.pnpbp.2019.02.015 [DOI] [PubMed] [Google Scholar]
  501. Zeeb FD, Floresco SB, & Winstanley CA (2010). Contributions of the orbitofrontal cortex to impulsive choice: interactions with basal levels of impulsivity, dopamine signalling, and reward-related cues. Psychopharmacology, 211(1), 87–98. 10.1007/s00213-010-1871-2 [DOI] [PubMed] [Google Scholar]
  502. Zeng M, Bai G, & Zhang M (2019). Anchoring high concentrations of SynGAP at postsynaptic densities via liquid-liquid phase separation. Small GTPases, 10(4), 296–304. 10.1080/21541248.2017.1320350 [DOI] [PMC free article] [PubMed] [Google Scholar]
  503. Zeng Z, Dong Y, Zou L, Xu D, Luo X, Chu T, Wang J, Ren Q, Liu Q, & Li X (2023). GluCEST imaging and structural alterations of the bilateral hippocampus in first-episode and early-onset major depression disorder. Journal of Magnetic Resonance Imaging, 58(5), 1431–1440. 10.1002/jmri.28651 [DOI] [PubMed] [Google Scholar]
  504. Zetzsche T, Preuss UW, Frodl T, Schmitt G, Seifert D, Münchhausen E, Tabrizi S, Leinsinger G, Born C, Reiser M, Möller HJ, & Meisenzahl EM (2007). Hippocampal volume reduction and history of aggressive behaviour in patients with borderline personality disorder. Psychiatry Research, 154(2), 157–170. 10.1016/j.pscychresns.2006.05.010 [DOI] [PubMed] [Google Scholar]
  505. Zhang Q, Chen X, Li S, Yao T, & Wu J (2021a). Association between the group III metabotropic glutamate receptor gene polymorphisms and attention-deficit/hyperactivity disorder and functional exploration of risk loci. Journal of Psychiatric Research, 132, 65–71. 10.1016/j.jpsychires.2020.09.035 [DOI] [PubMed] [Google Scholar]
  506. Zhang L, Lucassen PJ, Salta E, Verhaert PDEM, & Swaab DF (2022). Hippocampal neuropathology in suicide: gaps in our knowledge and opportunities for a breakthrough. Neuroscience and Biobehavioral Reviews, 132, 542–552. 10.1016/j.neubiorev.2021.12.023 [DOI] [PubMed] [Google Scholar]
  507. Zhang WH, Zhang JY, Holmes A, & Pan BX (2021b). Amygdala circuit substrates for stress adaptation and adversity. Biological Psychiatry, 89(9), 847–856. 10.1016/j.biopsych.2020.12.026 [DOI] [PubMed] [Google Scholar]
  508. Zhao Q, Che R, Zhang Z, Wang P, Li J, Li Y, Huang K, Tang W, Feng G, Lindpaintner K, He L, & Shi Y (2011). Positive association between GRIN2B gene and bipolar disorder in the Chinese Han Population. Psychiatry Research, 185(1–2), 290–292. 10.1016/j.psychres.2009.11.026 [DOI] [PubMed] [Google Scholar]
  509. Zhao J, Verwer RWH, Gao SF, Qi XR, Lucassen PJ, Kessels HW, & Swaab DF (2018). Prefrontal alterations in GABAergic and glutamatergic gene expression in relation to depression and suicide. Journal of Psychiatric Research, 102, 261–274. 10.1016/j.jpsychires.2018.04.020 [DOI] [PubMed] [Google Scholar]
  510. Zhou XT, Bao WD, Liu D, & Zhu LQ (2020). Targeting the neuronal activity of prefrontal cortex: new directions for the therapy of depression. Current Neuropharmacology, 18(4), 332–346. 10.2174/1570159X17666191101124017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  511. Zhou Q, Xiao M, & Nicoll RA (2001). Contribution of cytoskeleton to the internalization of AMPA receptors. Proceedings of the National Academy of Sciences of the United States of America, 98(3), 1261–1266. 10.1073/pnas.98.3.1261 [DOI] [PMC free article] [PubMed] [Google Scholar]
  512. Zimmermann AM, Jene T, Wolf M, Görlich A, Gurniak CB, Sassoè-Pognetto M, Witke W, Friauf E, & Rust MB (2015). Attention-deficit/hyperactivity disorder-like phenotype in a mouse model with impaired actin dynamics. Biological Psychiatry, 78(2), 95–106. 10.1016/j.biopsych.2014.03.011 [DOI] [PubMed] [Google Scholar]
  513. Zimmermann KS, Richardson R, & Baker KD (2019). Maturational changes in prefrontal and amygdala circuits in adolescence: implications for understanding fear inhibition during a vulnerable period of development. Brain Sciences, 9(3), 65. 10.3390/brainsci9030065 [DOI] [PMC free article] [PubMed] [Google Scholar]
  514. Zuo DY, Zhang YH, Cao Y, Wu CF, Tanaka M, & Wu YL (2006). Effect of acute and chronic MK-801 administration on extracellular glutamate and ascorbic acid release in the prefrontal cortex of freely moving mice on line with open-field behavior. Life Sciences, 78(19), 2172–2178. 10.1016/j.lfs.2005.09.022 [DOI] [PubMed] [Google Scholar]

RESOURCES