324
Views
23
CrossRef citations to date
0
Altmetric
Review

A selective review of glutamate pharmacological therapy in obsessive–compulsive and related disorders

, , &
Pages 115-131 | Published online: 28 Apr 2015

Abstract

Glutamate, an excitatory central nervous system neurotransmitter, is emerging as a potential alternative pharmacological treatment when compared to gamma-aminobutyric acid (GABA)-, dopamine-, and serotonin-modulating treatments for neuropsychiatric conditions. The pathophysiology, animal models, and clinical trials of glutamate modulation are explored in disorders with underlying inhibitory deficits (cognitive, motor, behavioral) including obsessive–compulsive disorder, attention deficit hyperactivity disorder, Tourette syndrome, trichotillomania, excoriation disorder, and nail biting. Obsessive–compulsive disorder, attention deficit hyperactivity disorder, and grooming disorders (trichotillomania and excoriation disorder) have emerging positive data, although only scarce controlled trials are available. The evidence is less supportive for the use of glutamate modulators in Tourette syndrome. Glutamate-modulating agents show promise in the treatment of disorders of inhibition.

Introduction

The use of glutamate-modulating drugs for neuropsychiatric conditions is emerging as an alternative to three decades of use of dopamine-, serotonin-, and gamma-aminobutyric acid (GABA)-modulating drugs in neuropsychiatry.Citation1Citation4 While such use is still nascent in clinical settings, there is promise of significant impact on improved outcomes for patients. Research in genetic epidemiology and neurophysiology is pinpointing glutamate, the essential excitatory central nervous system (CNS) neurotransmitter, as a direct target for pharmacologic manipulation. In this selective review, we present current drug trial data in disorders that exhibit features of a lack of inhibitory control, including cognitive (obsessive–compulsive disorder [OCD]), motor (Gilles de la Tourette syndrome [TS]), behavioral (attention deficit hyperactivity disorder [ADHD]), and grooming (trichotillomania [TTM], excoriation disorder [ExD]) domains.

Obsessive–compulsive disorder

OCD is characterized by the recurrence of unwelcome and intrusive ideations, urges, or images (obsessions), as well as repetitive, rigid behaviors (compulsions).Citation5 Obsessions can include fears related to contamination, religious, and sexual themes. Compulsions include behaviors such as washing, checking, counting, ordering, and hoarding. Obsessions and compulsions exist in symptom groupings or dimensions, with the four most commonly reported OCD symptom dimensions being: 1) contamination/washing; 2) aggressive/sexual/religious obsessions; 3) counting/repeating/checking compulsions; and 4) hoarding tendencies.Citation6 The prevalence rate of OCD in adults in the United States is 2.3%,Citation7 while it is lower in pediatric populations. In one study of 10,438 children in the United Kingdom, the prevalence rate of OCD was 0.25%.Citation8 Standard pharmacologic treatment consists of serotonin reuptake inhibitors (SRIs), with moderate efficacy in children. A primary treatment approach in children with OCD is cognitive behavioral therapy (CBT) with exposure to OCD-related fears (exposure–response prevention [ERP]). The exposure paradigm is thought to be dependent upon glutamate-facilitated extinction learning.Citation9 With medication and CBT/ERP, up to 54% of children with OCD experience clinical remission, but only 39% experience it with CBT alone and 21% with medication alone.Citation10

A developmentally-mediated brain dysplasia in cortico–subcortico–cortical networks has been postulated to underlie pediatric OCD.Citation11 Given that glutamate signaling is critical in early brain development through the facilitation of neuronal proliferation, migration, and differentiation, the formulation of the glutamate hypothesis of OCD relies on a dysregulation in developmental glutamate signaling homeostasis.Citation12 A rich expression of glutamate and GABA neurotransmitter activity in the fetal and postnatal brain supports early neuronal growth and maturation. Metabotropic glutamate receptor (mGluR)1, for example, has high levels of expression in the fetal brain, especially in the striatum, with subsequent progressive decreases, while mGluR4 is expressed mostly in the adult brain.Citation13 In a synchronized manner, both ionic and mGluRs are expressed in specific brain regions at specific times during brain development.Citation14 The excitatory effect of glutamate neuronal networks makes it important for effective efferent activity, but it also creates the potential for excitotoxicity.Citation15 In line with the glutamate hypothesis, functional imaging studies of OCD have shown an overactive cortico–striato–thalamo–cortical (CSTC) loop, with glutamate playing a central excitatory role (cortex, thalamus), while GABA and other inhibitory neurotransmitter systems are operant in the striatum and other subcortical regions.Citation16 Overactivity of the right caudate in OCD and the limbic CSTCCitation17,Citation18 is postulated to be a parallel dysfunction in glutamate homeostatic systems, such as the glutamate transporter family. Among members of this family of transporters, SLC1A1 in chromosome region 9p24, has emerged as an intriguing positional candidate gene for OCD. Multiple studies support the association of OCD with SLC1A1 variants,Citation19Citation21 including specific possible functional mutations,Citation22 but a more recent meta-analytic study has lent less support to its impact.Citation23 In the postsynaptic membrane, receptor conformation variants are plausibly linked to OCD. The NMDAR2B component of the N-methyl-D-aspartate (NMDA) receptor is codified by GRIN2B in chromosome region 12p12. GRIN2B variants have been linked to OCDCitation24,Citation25 and following the glutamate pathway’s downstream connections, variants in BDNF have also been linked to OCD. These BDNF variants include SNP rs2883187,Citation26 SNP rs1519480,Citation27 and the Met allele of Val66Met.Citation28 The evidence is thus mounting for a direct role of glutamate in the pathophysiology of OCD (); however, it is too early to determine whether glutamate dysregulation is a sign of intracortical excitability due to aberrant CSTC aberrant inputs,Citation29 another unknown glutamatergic abnormality, or a reactive effect of the disorder itself on brain function.Citation30

Table 1 Glutamate-modulating pharmacotherapy of obsessive–compulsive disorder

Animals have been heuristically useful in understanding the pathophysiologic mechanisms in OCD that may lead to drug discovery. The inhibition of marble-burying behavior has been used as a traditional model to test for anxiolytic drugs and OCD-related hoarding behaviors. In a test specific to anxiolytic effects, the mGluR5 (group 1) mediates marble-burying behavior in mice, and the mGluR5 antagonist, MPEP, is able to inhibit these behaviors.Citation31 Similarly, the marble-burying behaviors are also mediated by group 2 mGluRs (mGluR2 and mGlur3).Citation32 Additional support for glutamate-related animal models in OCD is provided by Sapap3 mutant mice, a knockout (KO) mouse that alters a synaptic density-associated protein. Sapap3 mutant mice demonstrate an excess of grooming behaviors, which is decreased significantly with the use of the selective SRI (SSRI) fluoxetine.Citation33 Additionally, in these KO mice, the surface expression and activity of mGluR5s are increased, suggesting that glutamate-driven overactive mGluR5 transmission unfavorably alters synaptic plasticity. In effect, a positive allosteric modulator of mGluR5 reproduces this effect in wild-type mice.Citation34 Interestingly, mGluR5 receptors have functional connections to BDNF, DLGAP1, and GRIN2B, as shown by a bioinformatics approach (http://www.string-db.org). In the rat attenuation model of OCD, decreasing rewards to lever pressing is used to identify those animals who “perseverate” in pressing behavior despite lower rewards. The excessive response can be abolished with fluoxetine and other serotonin-enhancing agents, but not by haloperidol or benzodiazepines.Citation35 In a follow-up paradigm, the NMDA agonist D-cycloserine (DCS), but not an NMDA antagonist, helped attenuate the perseverative response, supporting a role for NMDA-facilitated learning of the extinction process.Citation36

Clinical trials of glutamate-modulating drugs in OCD

Amantadine

Amantadine (1-adamantanamine hydrochloride) is an antiviral drug that was formerly, but no longer, used for influenza prophylaxis.Citation37 Its current uses are to improve alertness and arousal in post-traumatic brain injury in children,Citation38 improve executive dysfunction in patients with Alzheimer’s dementia,Citation39 and treat the early stages of Parkinson’s.Citation40 The mechanism of action is thought to consist of releasing dopamine from the presynapse, in addition to possible negative modulation of the NMDA receptor.Citation41 While it can be neuroprotective due to its glutamate antagonism properties, amantadine has also resulted in acute neuropsychiatric side effects such as hallucinations and confusion.Citation42 A case study reported that a treatment-refractory patient with OCD responded to amantadine (200 mg/day) added to clomipramine (225 mg/day).Citation43 In an open-label study of amantadine in eight patients with OCD who had failed one SSRI trial,Citation44 Yale–Brown Obsessive Compulsive Scale (Y-BOCS) scores improved for compulsions (15.3±3.2 versus 10.6±4.7; P<0.02; degrees of freedom [df] =7; t=2.36) and obsessions (12.7±3.3 versus 8.1±5; P<0.05; df=7; t=2.36).

D-cycloserine

DCS is an analog of D-alanine and an NMDA receptor partial agonist at the glycine-binding site. It has been well established that DCS has the ability to contribute to fear extinction learning.Citation45 DCS facilitates conditioned fear extinction in rats when injected in the amygdala or systemically, suggesting that treatments that activate NMDA receptors can promote the learning of extinction learning.Citation46 The sum of preclinical studies thus supports the blocking of extinction learning by NMDA antagonistsCitation47 and its facilitation by agonists such as DCS.Citation48 In the treatment of OCD, DCS has been combined with ERP as a treatment option for nonresponders to traditional ERP. DCS administered 2 hours prior to ERP can produce an accelerated decrease in obsession-related fear ratings in comparison to the placebo across four ERP therapy sessions.Citation49 This acceleration of response was also seen in another study of 16 adults with OCD, nine of whom received DCS + ERP, while seven received placebo. Midtreatment, the active group had significantly lower Y-BOCS scores (t=2.87; df=21; P=0.009) with a large effect size (Cohen’s d =1.17). At 1-month follow-up, differences were no longer apparent.Citation50 In children with OCD, a negative study suggested that the differences in methodology may affect results.Citation51 A meta-analysis of 13 studies, only one using pediatric OCD subjects, showed a small-to-moderate effect size for the use of DCS augmentation for exposure therapy (Cohen’s d =−0.34).Citation52

Glycine

Glycine is biosynthesized from serine in a reversible folate-dependent reaction to conform a nonessential amino acid (NH2CH2COOH), which can act as a neurotransmitter.Citation53 In the nervous system, glycine is inhibitory in the spinal cord, playing a role in inhibitory postsynaptic potentials, but excitatory in the brain and cerebellum, where it is a glutamate coagonist at the NMDA receptors.Citation54 Originally studied as a neurotransmitter for pain signaling in the spinal cord, it has gained importance in CNS disorders due to its NMDA coagonist properties and its possible role in determining the number and positioning of cortical interneurons. For example, activation of the NMDAR D-serine/glycine site or inhibition of glycine transport has been pharmacologically induced to benefit animal models and clinical trials in schizophrenia.Citation55 In a placebo-controlled double-blind study of OCD,Citation56 24 subjects were assigned glycine 60 g/day or placebo. Glycine noncompliance, partly due a lack of palatability of the compound, led to ten dropouts. Follow-ups at 4-weeks, 8-weeks, and 12-weeks showed a marginally greater decrease of Y-BOCS OCD severity (P=0.053) in the glycine group (6.04 mean decrease; number [n]=5) compared to the placebo group (1.0 mean decrease; n=9). There were two out of five responders in the glycine group and zero out of nine responders in the placebo group (P=0.11).Citation56

Ketamine

Ketamine (2-[2-chlorophenyl]-2-[methylamino] cyclohexanone) is a noncompetitive, nonselective, high-affinity NMDA receptor antagonist with psychotomimetic properties. Related to the more potent phencyclidine, its profile additionally includes a short half-life and weak binding to the mu opiate receptor, monoamine transporter sites, and acetylcholinesterase receptors.Citation57 Based on preclinical research and the emerging glutamate hypothesis of mental illness, ketamine was used at 0.5 mg/kg to treat refractory depression with positive results.Citation58 More recent overviews of this use of ketamine overall confirm its utility, but unanswered questions still remain about the precise mechanism of action and long-term benefits.Citation59

The treatment of OCD with ketamine is in the initial stages of validation. An isolated case report used two intravenous (IV) ketamine infusions (0.5 mg/kg) 1 week apart on a 24-year-old female with OCD. No symptom reduction was observed after placebo; on ketamine infusion, almost complete reduction of obsessions was observed up to 7 days postinjection.Citation60 In an open-label trial of IV ketamine 0.5 mg/kg in ten patients with refractory OCD, the initial symptom reduction of OCD symptoms was observed, along with decreases in the Hamilton Depression Rating Scale-17, a clinician-administered Dissociative States Scale, and the Clinical Global Impression (CGI) scale. Though the decrease in symptoms was maintained at 50% in four out of seven depressed patients, none of the OCD patients (n=10) sustained significant improvements.Citation61 In a recent randomized, double-blind, placebo crossover study, adults with OCD (n=15) received two IV ketamine infusion treatments. The comparison groups were saline (n=7) and low-dose ketamine (0.5 mg/kg) (n=8). Those in the full-dose experimental group reported substantial improvements in OCD symptom severity, with 50% meeting the criteria for treatment response (≥35% Y-BOCS reduction) using the OCD visual analog scale and the Y-BOCS. The residual effects of ketamine lasted longer than the expected 1-week postinfusion.Citation62

Lamotrigine

Lamotrigine (6-[2,3-dichlorophenyl]-1,2,4-triazine-3-,5-diamine) is an anticonvulsant, antiepileptic drug with voltage-gated Na-channel blocking properties.Citation63 In addition, lamotrigine also facilitates GABA release and reduces the presynaptic release of glutamate.Citation64 Its anticonvulsant and mood-stabilizing properties make it a treatment of choice for the management of seizuresCitation65 and manic–depressive illness, in particular through putative neuroprotective effects.Citation66 It is especially effective in the prevention of the depressive component of bipolar disorder,Citation67 bipolar relapse,Citation68 and unipolar depression resistant to treatment.Citation69 One double-blind trial has also shown preliminary efficacy in the treatment of the re-experiencing and numbing symptoms of post-traumatic stress disorder.Citation70

The glutamate-modulating properties of lamotrigine, based on the property of inhibiting Na+ channel-opening glutamate release,Citation71 suggests that it may have some utility in the treatment of OCD. In a single case study of a 59-year-old woman with treatment-resistant OCD,Citation72 the combination of a stable dose of clomipramine (225 mg/day) and lamotrigine (up to 150 mg/day) yielded a significant reduction in OCD Y-BOCS severity scores after 10 weeks. A subsequent 16-week, double-blind, placebo-controlled study of lamotrigine in 33 SSRI-resistant OCD patients showed that lamotrigine, added to standard SSRI treatment, substantially improved obsession and compulsion severity (P<0.0001).Citation73

Memantine

Memantine (3,5-dimethyladamantan-1-amine) is a noncompetitive NMDA receptor antagonist with neuroprotective properties in cortical neuron culturesCitation74 and in humans.Citation75 These properties propelled its use in Parkinson’s disease,Citation76 Huntington’s disease,Citation77 and Alzheimer’s disease.Citation78 The same glutamate-modulating property has motivated the assessment of memantine in OCD.

A reported case study of a 15-year-old female with severe OCD used memantine 5 mg twice daily (bid) in addition to preexisting citalopram. Obsessions and rituals were soon controlled for the first time in 9 months.Citation79 An additional case study yielded similar results in a 29-year-old male, wherein 3 weeks of 15 mg/day of memantine produced over a 40% reduction in the Y-BOCS severity score, although another patient did not benefit.Citation80 Next, an open-label trial enrolled 15 SRI-resistant OCD patients for a 12-week augmentation using memantine 10 mg bid.Citation81 Data from 14 of the 15 subjects yielded significantly lower Y-BOCS scores compared to baseline (P<0.05) and 42.9% of the participants were classified as responders (≥25% reduction in Y-BOCS scores and a CGI rating of “much” or “very much” improved).Citation81 Another open-label trialCitation82 in ten OCD and seven generalized anxiety disorder subjects also used memantine 10 mg bid as a standalone or add-on psychopharmacological treatment. Patients with OCD experienced a 41% reduction of Y-BOCS severity scores compared to baseline (P<0.001), with three of ten subjects classified as responders.Citation82 Finally, an 8-week randomized, double-blind, placebo-controlled study of memantine 10 mg bid was conducted on 42 patients with moderate-to-severe OCD.Citation83 Of 38 completers, memantine was titrated to 10 mg bid. By 8 weeks, 100% of those on memantine and 32% of those on placebo achieved partial or complete response (P<0.001).Citation83

N-acetylcysteine

N-acetylcysteine (NAC), a derivative of cysteine, has been used as a mucolytic in bronchitisCitation84,Citation85 and as an anti-inflammatory antioxidant.Citation86 Administration of NAC within 16 hours postingestion is a primary treatment of acute acetaminophen toxicity;Citation87 it can also be useful in the management of toxicity when hepatic failure has been established.Citation88,Citation89 Based on its ability to replenish the endogenous active antioxidant glutathione and direct scavenging of reactive oxygen species, NAC has been targeted to decrease doxorubicin cardiotoxicity in rat animal modelsCitation90 and the oxidative damage in acute respiratory distress syndrome.Citation91 Its thiol properties have supported its use in the protection of chemotherapy-induced cytotoxicity, if administered shortly after cisplatin,Citation92 and its microcirculatory effects and inhibition of neutrophil aggregation in ischemia–reperfusion cardiac injury for coronary artery bypass graft surgery.Citation93 Finally, even dietary NAC can replenish the antioxidant glycine pool, with a resultant increase in cysteine, glutamine, and oxidized glutathione.Citation94

In a single study that evaluated the total oxidant and antioxidant status in serum, there was increased oxidant status and lower antioxidant status, with a resulting high oxidative stress index in children with OCD.Citation95 Another study in adults with OCD measured erythrocyte malondialdehyde, a product of lipid peroxidation, and found that OCD was associated with higher levels of malondialdehyde, suggesting that a higher oxidation level was present. In this study, the antioxidant vitamin E, but not vitamin C, was also significantly lower in patients with OCD.Citation96 Clinically, only a single case study of a 58-year-old woman with SRI-refractory OCD, in whom 3 g/day of NAC augmented fluvoxamine, reported a decrease of her Y-BOCS score from 32 (severe OCD) to 9 (no OCD) over 12 weeks.Citation97

Riluzole

Riluzole (2-amino-6-trifluoromethoxy benzothiazole) is a negative regulator of CNS glutamate activity.Citation98 Animal models suggest that riluzole is not specific to glutamate – it also decreases the release of acetylcholine and dopamine-trough mechanisms independent of NMDA or AMPA receptors.Citation99 Other cell experiments support that in cortical neurons, riluzole inhibits voltage-dependent Na+ channels, resulting in nonglutamate-mediated anticonvulsant properties.Citation100 The neuroprotective properties of riluzole made it an attractive option for the treatment of amyotrophic lateral sclerosis (ALS), a neurodegenerative disease of cortical, brainstem, and spinal cord motor neurons.Citation101 Based on successful double-blind trials, in 1996, riluzole was approved for the treatment of ALS.Citation102 Clinical experience with riluzole in ALS suggests that it prolongs survival,Citation103 along with an adequate long-term use safety profile.Citation104

Riluzole has been used in open and randomized clinical trials for the management of OCD symptoms. An early open-label study on 13 treatment-resistant patients with OCD ages 18–65 years used riluzole 50 mg bid to augment therapy. Of the 13 patients, seven out of 13 (54%) reported a >35% reduction in the Y-BOCS severity score. Of these, five out of 13 (38%) were treatment responders. Depression and anxiety measures also improved, and no concerning side effects were reported.Citation105 Another open-label study of six children with OCD demonstrated that four out of six (67%) had a mean 40% decrease in OCD symptom severity.Citation106 However, a recent double-blind randomized trial on 60 children with OCD, using riluzole 100 mg/day for augmentation, did not demonstrate an advantage over placebo.Citation107

Topiramate

Topiramate (C12H21NO8S) is an anticonvulsant agent with linear kinetics and low binding to plasma proteins.Citation108 Its glutamate-modulating properties are mediated through AMPA receptor antagonism, in addition to the blockage of voltage-dependent Na+ channel and the potentiation of GABA-A neurotransmission.Citation109 It also inhibits AMPA and kainate receptor facilitation of Ca2+ influx, thus providing a potential buffer to glutamatergic excitotoxicity.Citation110 Topiramate’s anorectic properties have motivated its use for weight control in combination with phentermine,Citation111 and its pain control properties support topiramate’s use in the prophylaxis of chronic migraine.Citation112

A case report of a 45-year-old paroxetine-resistant patient with OCD highlighted the use of topiramate 150 mg/day.Citation113 After 9 weeks, the Y-BOCS OCD severity score decreased significantly.Citation113 A more extended review of 16 consecutive adult patients with OCD, who had only partial response to SRI with or without neuroleptic augmentation, were reviewed after topiramate was added. Upon topiramate addition at 250 mg/day over an average of 9 weeks, there was a positive response in eleven out of 16 (69%) patients.Citation114 A 12-week, double-blind, placebo-controlled trial of topiramate was conducted in 36 patients with OCD.Citation115 Eighteen patients received a 12-week SSRI trial in addition to topiramate (mean dose: 180 mg); the other 18 patients received placebo in addition to the SSRI. Only the compulsions subscale improved (P=0.01), not the obsessions or total Y-BOCS scales. However, many patients had discontinued treatment due to side effects in the active arm (5/18; 28%).Citation115 One case study of OCD emerging after topiramate treatment remains as an isolated report.Citation116

Pregabalin

Pregabalin, (S)-(+)-3-aminomethyl-5-methylhexanoic, was designed as a blood–brain barrier-permeable GABA analog related to gabapentin. Originally designed as an anticonvulsant, it has been found to be effective in the treatment of hyperalgesia and pathologic anxiety.Citation117 In hippocampal neurons, pregabalin decreases exocytosis, which opens neurotransmitter vesicles from presynaptic sites, a process that appears to depend on NMDA activation.Citation118 Based on its anxiolytic properties, pregabalin was used in an 8-week open-label trial in ten OCD SRI–neuroleptic-resistant patients.Citation119 Adjunctive pregabalin was administered at 225–675 mg/day, resulting in a significant reduction of OCD Y-BOCS severity scores from 27.1 (standard deviation [SD] =6.5) to 12.3 (SD =7.1). Overall, a 35% symptom improvement was seen in eight of the patients’ Y-BOCS scores.Citation119

Tourette syndrome

TS is a childhood onset neurobehavioral disorder characterized by >12 months of multiple motor tics and at least one vocal tic during the course of the disorder. Tics are stereotyped, suppressible, and suggestible repetitive movements of single muscle groups (simple tics) or multiple muscle groups (complex tics).Citation120 Involuntary and detrimental to daily functioning, tics are commonly treated with behavioral therapy with an emphasis on tic suppressionCitation121 to improve psychiatric and psychosocial functioning in childrenCitation122 and adults.Citation123 Clinically, TS is often comorbid with ADHD,Citation124 OCD,Citation125 as well as anxiety and mood disorders. Comorbidities significantly impact the quality of life of TS patients.Citation126

Pathophysiologically, TS has been associated with the CSTC with a key focus on synaptic neurotransmission abnormalities.Citation127,Citation128 One of the main proposed mechanisms in TS is the increased transmission of dopamine,Citation129 possibly due to abnormally high dopamine receptor prefrontal density.Citation130 An alternative view is illustrated by the putative involvement of glutamate in neurotransmission in CSTC circuits,Citation131 extensive interaction of dopamine and glutamate systems (),Citation132 and postmortem brain studies.Citation133

Table 2 Glutamate-modulating treatments for Tourette syndrome

Animal models in TS have been fraught with the innate complexity of modeling specific motor movements (tics) in nonhuman species. A working animal model will depend on the physiological assumptions about tics; for example, deficient gating has been proposed as a mechanism for propensity to tic expression based on premonitory urges associated with tics. The prepulse inhibition (PPI) of the acoustic startle reflex-deficit animal model is used to demonstrate deficient gating, and although not specific to TS, it can be used to explore pharmacologic options for tics.Citation134 An alternate animal model relies on D1 receptor variants that drive glutamatergic neurocircuitry excitability; a mouse model displays compulsive-like repetitive behaviors.Citation135,Citation136 Interestingly, in these animal models, PPI is disrupted by the chemically-induced antagonism of D1 receptors in the medial prefrontal cortex (dopamine mechanism) and NMDA receptors in the ventral hippocampus (glutamate mechanism).Citation137 These models suggest complex interactions in the cortical–subcortical neurocircuitry that involve dopamine and glutamate influences, along with a decrease in PPI.

Clinical trials of glutamate-modulating drugs in TS

Ketamine

Ketamine, a high-affinity NMDA receptor antagonist, has been found to increase PPI levels, making it a plausible drug to ameliorate tics. No direct trials of ketamine in TS are available; therefore, data on the effect of ketamine on PPI are reviewed. To measure the cognitive and PPI effects of ketamine, 20 ketamine-naïve subjects with negligible baseline startle responses were given either a placebo or a dose of ketamine of 0.23 mg/kg over 5 hours, followed by ketamine 0.5 mg/kg over 1 hour. A significant increase in PPI levels was detected in the active drug group.Citation138 In another study with 16 healthy patients, researchers found that subanesthetic doses of ketamine significantly increased PPI.Citation139 In both trials, experienced cognitive effects resembled those seen in schizophrenia, but PPI levels were increased by ketamine rather than decreased, as seen in schizophrenic patients, suggesting utility in disorders with abnormal levels of PPI.

Lamotrigine

The efficacy of lamotrigine in TS has been variable. A single case study reported decreased TS symptoms in a 26-year-old woman with a clinical diagnosis of bipolar II disorder, TS, migraine, and complex partial seizures.Citation140 An initial Yale Global Tic Severity Scale (YGTSS) score of 39/50 was decreased over 6 months after a 12-week titration of lamotrigine to 300 mg/day.Citation140 Another case report documented increased tics after lamotrigine 200 mg/day for mood stabilization in a 55-year-old female with bipolar disorder.Citation141 Tics included shrugging her right shoulder, wagging her hips, pawing her feet on the ground, and vocal tics. After tapering lamotrigine dosage, her tic symptoms faded and disappeared in 2 weeks’ time.Citation141 TS symptom emergence, which improved on lamotrigine withdrawal, has been reported in five children using lamotrigine for the control of seizure disorders.Citation142,Citation143

Pregabalin

Motor tics that accompany TS are self-injurious ∼4%–43% of the time.Citation144 In one case study, a 54-year-old man with TS with self-injury presented with fibromyalgia, comorbid OCD, and self-injurious behaviors (tongue self-mutilation).Citation145 The patient was resistant to SSRIs, clomipramine, and haloperidol. Pregabalin monotherapy 600 mg/day was administered given the history of fibromyalgia and failure of other approaches; it was successful in improving self-injurious behaviors over 52 weeks of use.Citation145

Topiramate

A retrospective chart reviewCitation146 of 41 patients with TS who were administered topiramate (mean dose: 150 mg/day) for an average of 9 months, suggested that this drug could be effective for tic management. In this case series, topiramate was used as the primary monotherapy in 21 of 41 patients, while concomitant medications included neuroleptics (11/41), clonidine or guanfacine (6/41), tetrabenazine (3/41), and botox (3/41). Over 75% of subjects had “moderate to marked improvement” of tics. The main adverse effects were those common to topiramate: cognitive–language problems (25%) and aggression/mood swings (10%).Citation146 A single 10-week, randomized, double-blind, placebo-controlled, parallel group study compared topiramate versus placebo in 29 patients with TS.Citation147 The main outcome, “total tic score”, significantly improved in the active drug group (14.3 [SD =10.5]) compared to placebo (5.0 [SD =9.9]) (P<0.05). YGTSS motor, vocal, and total tic scores were significantly improved in the active drug group compared to placebo. No difference in adverse events was noted between the two groups.Citation147 To date, topiramate constitutes a second-line agent in the management of clinically impairing tics.

Attention-deficit hyperactivity disorder

ADHD is a neurobehavioral disorder characterized by clinically impairing inattention and/or hyperactivity–impulsivity, which arise during early development. ADHD is categorized into three subtypes: 1) predominantly inattentive; 2) predominantly hyperactive–impulsive; and 3) combined.Citation5 However, the full characterization of the syndrome should also emphasize related core deficits: an inability to inhibit the prepotent response;Citation148,Citation149 working memory developmental lags;Citation150 and executive dysfunction.Citation151 In a 2011 US survey, up to 11% of children and adolescents 4–17 years of age were affected, and less than two-thirds of diagnosed children were taking medication.Citation152

Anatomical abnormalities in ADHD include reductions in total cerebral, prefrontal cortex, basal ganglia, dorsal anterior cingulate cortex, corpus callosum, and cerebellar volumes.Citation153Citation155 Functional magnetic resonance imaging (fMRI),Citation156,Citation157 magnetic resonance spectroscopy (MRS),Citation158 and resting-state fMRI also detect abnormalities in ADHD when compared to normotypicals.Citation159 Specific to glutamate, a proton MRS ([1H]MRS) study of 40 adults with ADHD (24 medication-naïve), found that glutamate + glutamine (Glx) was lower in the basal ganglia and dorsolateral prefrontal cortex when compared to a control region (medial parietal cortex). Medication status did not alter the results, and the level of symptoms significantly correlated with Glx levels in the basal ganglia.Citation160 Adults with autism spectrum disorder have also displayed decreased Glx concentrations in the basal ganglia compared to a parietal cortex control region.Citation161 Other target regions in adults with ADHD, the right anterior cingulate cortex (decreased Glx)Citation162 and the left cerebellar hemisphere (increased Glx),Citation163 also have alterations in Glx (glutamate/glutamine) resonance. An additional study in children with ADHD found that particularly Glx – and not N-acetylaspartate, creatine/phosphocreatine (Cr), or choline – was increased in the right prefrontal cortex and left striatum.Citation164 An increased Glx ratio was seen in the right prefrontal cingulate in another study of patients with ADHD aged 8–54 years compared to controls.Citation165 The alterations in the Glx balance in different brain regions in ADHD are still of unknown valence. Although ADHD is known to be highly influenced by dopamine-linked neurocircuitry,Citation159 extensive interactions between the glutamate and dopamine neural systems support a role for glutamate in ADHD pathophysiology ().Citation148,Citation149 For example, in the prefrontal cortex, D1 and D2 receptors modulate glutamate transmission at the NMDA and AMPA receptor sites. In turn, the downregulation of glutamate transmission for NMDA receptors is affected by GABAergic interneurons via D1 receptors.Citation166 Following the demonstration of increased Glx content in ADHD,Citation167 the effect of medication on brain regions has been examined. Methylphenidate and atomoxetine effect a 56% reduction in Glx in the left striatum in parallel to decreasing ADHD symptoms.Citation168 In an analogous subsequent study in 13 treatment-naïve children with ADHD compared to controls, both Glx and Cr were increased pretreatment in the left striatum.Citation169 After methylphenidate treatment, symptoms were improved and only striatal creatine was found to be reduced.Citation169 A more recent MRS study in adolescents with ADHD found increased ratios of Glutamine/Inositol, Glutamate/Inositol, and Glx/Ino in the anterior cingulate – a brain region targeted for its role in attention in relation to error detectionCitation170 – in subjects with ADHD compared to controls.Citation171 However, these differences were not statistically significant. Additionally, no significant differences were seen in these metabolite ratios pre- and post-treatment with a methylphenidate long-acting preparation.Citation171

Table 3 Glutamate-modulating pharmacotherapy for attention deficit hyperactivity disorder

Animal models have been instrumental in understanding the biological underpinnings of ADHD, as well as the interaction of the dopamine and glutamate systems influencing attentional and hyperactivity behavioral domains. An early model located the dopamine receptor (DR)D1 as a potential target for genetic manipulation. The DRD1 KO mouse exhibits excessive motor activity, as well as reduced striatum dynorphin.Citation172 Later experiments reaffirm that DRD1 plays a role in attentional performance with D1 agonists improving, and D1 antagonists worsening, attentional performance in mouse models. D2 antagonists have no effect on attentional performance in these models.Citation173 A direct measure of the glutamate-stimulated release of dopamine in key ADHD-related brain regions used the spontaneously hypertensive rat model of ADHD (SHR). Compared to control rats, SHR showed increased glutamate-stimulated dopamine release in the substantia nigra, implicating altered glutamate regulation of dopamine neurons in this ADHD rodent model.Citation174 A recent extension of these findings confirmed increased evoked glutamate release in the cingulate and infralimbic cortices, as well as in the striatum, using the SHR model of ADHD.Citation175

Genetic lines of research are also pointing to the influence of glutamate in ADHD. A recent genome-wide study implicates glutamatergic synaptic adhesion molecules and mGluRs as etiologic in ADHD.Citation176 Another genome-wide study using copy-number variants identified copy-number variants spanning GRM genes.Citation177 DRD4 variants have also been associated with ADHD risk.Citation159 It had been shown earlier that DRD4-deficient mice display cortical hyperexcitability, suggesting that DRD4 stimulation negatively regulates glutamate activity, thereby curbing neurotoxicity.Citation178 In addition, manipulation of DRD4 activity in pyramidal cortical neurons resulted in reversible decreases of NMDA receptor-mediated currents, in association with inhibition of protein kinase A (PKA), activation of protein phosphatase 1, and inhibition of calmodulin-dependent protein kinase II (CAMKII).Citation179 Conversely, in DRD4-deficient mice, there was increased expression of DRD1 and NMDA receptors in the nucleus accumbens and hippocampal CA1 tissue, suggesting a functional relationship between DRD4, DRD1, and NMDA receptors.Citation180 These relationships carry over into the striatum, following more recent experiments with DRD4-deficient mice, which show that resting extracellular levels of glutamate are increased in the striatum, and glutamate clearance kinetics are significantly decreased in the dorsal striatum.Citation181 PKA, an intracellular target of DRD1 stimulation, is also implicated in attentional performance. Inhibition of PKA both reduces attentional performance and increases locomotor activity. In turn, PKA activates cyclic adenosine monophosphate (cAMP) responsive element binding protein (CREB), a transcription factor that binds to DNA “responsive elements” intracellularly, increasing the transcription of c-Fos, BDNF, somatostatin, VGF, enkephalin, and corticotrophin releasing hormone, among others.Citation182 Inhibition of PKA reduces the accuracy of a serial reaction time task in rodents, a test that parallels the continuous performance test in humans. The same effect is observed with the disruption of CREB.Citation165 The interplay of glutamate signaling and CREB influences attentional and locomotor systems in animal models. Among these models, glutamate homeostasis in the ventromedial prefrontal cortex (vmPFC) is an important framework for exploring the symptoms of ADHD. Infusion of rat vmPFC with the NMDA antagonist 3-(R)-2-carboxypiperazin-4-propyl-1-phosphonic acid ([R]-CPP) decreases attentional performance, while increasing impulsivity and perseverative responses. These changes are accompanied by increased excitotoxic glutamate efflux in the vmPFC and changes in CREB expression in the vmPFC (increased) and striatum (decreased). Counteraction of the glutamate imbalance produced by (R)-CPP is achieved with the mGluR2–3 agonist agent, LY379268, which restores attentional performance and impulsivity, but not perseverative responses. This model suggests that the NMDA and mGluR2–3 pathways may underlie the core components of ADHD.Citation183 Another rodent model of ADHD is the Naples high-excitability rat, which is bred to display behavioral arousal to novelty.Citation184 Forebrain levels of glutamate, as well as d- and l-aspartate, are elevated in Naples high-excitability rats, suggesting that these levels may be causing neurotoxicity in the frontostriatal pathways.Citation185

Clinical trials of glutamate-modulating drugs in ADHD

Amantadine

Recent trials of amantadine in pediatric populations have included its use in autism and ADHD.Citation41 In an early open-label trial of amantadine, eight children with ADHD 10–13 years of age were taken off stimulant medication and provided with amantadine 100 mg bid for 1 month. While two children had similar responses as they had to methylphenidate, most only had an intermediate response compared to methylphenidate.Citation186 A 6-week, open-label trialCitation187 using amantadine 50–150 mg on 24 stimulant-naïve children with ADHD aged 5–13 years showed superiority over placebo. Only one child could not complete the trial due to headaches, and another had a transient decrease in appetite. Up to 60% of children had a >25% response in parent ADHD ratings, and 50% by teacher ratings.Citation187 A head-to-head 6-week comparison trialCitation188 of amantadine (100–150 mg/day) versus methylphenidate (20–30 mg/day) among 28 boys and 12 girls with ADHD aged 6–14 years showed no difference between the two drugs. Parent ratings showed 55% were responders on methylphenidate and 50% on amantadine; the teacher ratings were 35% responders for methylphenidate and 30% for amantadine.Citation188 From these trials, amantadine has a modest effect in treating ADHD. Given its primary glutamate-modulating action, amantadine may be useful in treating symptoms of ADHD in specific subgroups of affected children.

Lamotrigine

A retrospective case series of 40 adult patients aged 16–55 years with ADHD comorbid with bipolar II or recurrent depression was conducted to gauge the effect on ADHD symptoms.Citation189 Most patients (38/40) were on methylphenidate as well. With lamotrigine doses ranging from 25–250 mg, CGI scores were improved over the 48-month follow-up span.Citation189 Antiepileptics can worsen attentional performance, and surveys can been conducted to assess these detrimental effects. In one survey on newly diagnosed childhood absence epilepsy, valproate, but not lamotrigine or ethosuximide, worsened attentional performance.Citation152 Overall, lamotrigine improved cognition and attention in 24 of 45 children receiving lamotrigine for seizure control in one study,Citation190 and the improvement of ADHD symptoms with lamotrigine does not appear to be dependent on ameliorating seizure activity.Citation158 Given its benign to favorable cognitive profile, clinicians have advocated using lamotrigine as the antiepilepitc of choice when seizures are comorbid with ADHD.Citation191 Currently, only indirect evidence supports the use of lamotrigine for the management of ADHD symptoms in children.

Memantine

Memantine is related to amantadine, with both increasing striatal dopamine via NMDA antagonism.Citation192 Memantine 20 mg/day was compared to placebo in 16 children ages 6–12 years to test for efficacy in ADHD. In this pilot study, memantine was found to be efficacious, with no significant adverse effects.Citation193 A more recent open-label 12-week trialCitation194 on 28 adults with ADHD or ADHD not otherwise specified used memantine 10 mg bid. Exclusion criteria decreased the subject pool to 28 participants. The Adult ADHD Investigator Symptom Report yielded a reduction in total symptoms (–17.5; P<0.001), inattentive symptoms (–10.6; P<0.001), and hyperactive symptoms (–6.9; P<0.01) at the 12-week assessment. A total of 44% of participants had CGI ratings of much or very much improved.Citation194

N-acetylcysteine

A trial of NAC was conducted in 49 adults with systemic lupus erythematosis over 3 months.Citation195 Using an ADHD rating scale, patients with systemic lupus erythematosis had significantly greater ADHD symptoms than controls. The ADHD symptoms significantly improved on NAC 2.4–4.8 g/day (P<0.001).Citation195

Trichotillomania, excoriation disorder (OCD-related disorders)

TTM is an OCD-related disorder (OCD-RD) characterized by repetitive hair pulling resulting in noticeable hair loss. Individuals may develop rituals around the selection and removal of hair, and may engage in manipulations like rolling, examining, biting, or swallowing the hair after removal.Citation164 Children with TTM engage in automatic hair pulling, while older children appear to have more focused pulling and are more aware of associated urges, with increased frequency of hair pulling.Citation154

Dermatillomania or ExD is characterized by recurrent skin picking resulting in skin lesions. The age at onset can be in childhood and triggers include stress, boredom, or the feel or look of the skin.Citation157 A striking observation is the high degree of comorbidity associated with skin-picking behaviors, including OCD, body dysmorphic disorder, substance use disorders, eating disorders, TTM, kleptomania, and compulsive buying.Citation196 The prevalence of skin-picking disorder ranges from 1.4%–5.4% based on two recent community samples.Citation197,Citation198

TTM and ExD are classified by the Diagnostic and Statistical Manual of Mental Disorders, 5th edition, as OCD-RD. These body-focused repetitive behaviors may be preceded by a sense of increasing tension and followed by a sense of gratification, pleasure, or relief. A continuum of conscious awareness may be associated with TTM and ExD, and the behaviors may be preceded by stress, anxiety, or boredom.Citation151

The pathophysiology of OCD-RD has been investigated; however, much work needs to be done to further clarify their relationship to OCD. There are indications of increased familiality in TTMCitation199 and ExD,Citation200 suggesting a genetic diathesis. A lack of motor inhibitory control, such as in the Go–NoGo task in TTMCitation201 and in the stop–signal task in ExD,Citation160 point to an underlying neural inhibitory network deficit. Positron emission tomography studies show an increase in mean global metabolism in TTM compared to controls, as well as normalization of increased orbitofrontal and anterior cingulate regions with clomipramine treatment.Citation202 More recently, and paralleling neuropsychological inhibitory deficit findings, a white matter tract study of subjects with TTM found reduced fractional anisotropy (white matter disorganization) in the anterior cingulate, pre-supplementary motor area, and temporal cortices,Citation161 as well as increased mean diffusivity in the fronto–striato–thalamic white matter tracts.Citation203 In ExD, analogous white matter disorganization is also found in areas associated with inhibitory control, such as the tracts emerging from the inferior right prefrontal regions.Citation204 These pathophysiologic changes suggest a neurocircuitry imbalance in inhibitory neural control systems ().

Table 4 Glutamate-modulating pharmacotherapy in grooming disorders and OCD-related disorders

Animal models of OCD-RD have sought to simulate repetitive behaviors that include animal self-grooming motor sequences. Hox genes are developmental homeobox genes that direct somatic body structure,Citation153 one of which is hoxb8, a transcription factor directing anterior limb development.Citation205 The hoxb8 mutant mouse exhibits an abnormally high levels of grooming behaviors, causing skin lesions and hair removal.Citation206 Another notable example of a mouse model of grooming behaviors is the SAP90/PSD95-associated protein 3 (SAPAP3) KO mouse, which exhibits heightened anxiety-like behaviors and excessive self-grooming behaviors leading to facial hair loss and skin lesions.Citation33 SAPAPs are SAP90/PSD95-associated proteins that act as synaptic linker proteins between glutamate receptor-binding proteins and the cytoskeleton, while SAPAP3 is particularly highly expressed in glutamatergic synapses of the striatum.Citation207 In humans, SAPAP3 is expressed in dendrites, and SAPAP3 genetic variants have been associated with TTM and ExD.Citation208,Citation209

Clinical trials of glutamate-modulating drugs in TTM and ExD

N-acetylycysteine

A 12-week, double-blind, placebo-controlled trial of NAC 1.2–2.4 g/day in 50 adults with TTM showed a greater reduction in hair-pulling behaviors in the active drug arm.Citation163 Up to 56% of patients taking NAC were much or very much improved, while only 16% of subjects receiving placebo had the same benefit. NAC produced no adverse events in the treated group.Citation163 Following the randomized trial, additional case reports of NAC in TTM have been reported. A case report of two females aged 23 years and 19 years showed benefit of NAC 1,200 mg/day, with complete hair regrowth at 3 months and 2 months, respectively, after long courses of illness.Citation156 Another case report of a 58-year-old female with TTM used NAC 1,200 mg/day, with a gradual improvement of hair loss observed over a 10-week period.Citation150 In contrast to positive trials in adults, a double-blind placebo randomized trial of NAC in TTM, which included 39 children ages 8–17 years, showed no difference between NAC (25% improved) when compared to placebo (21% improved).Citation155

The use of NAC in ExD is highlighted by a case report of five patients with TTM or ExD who were given NAC for symptom management. One of the reported patients had ExD, which responded to NAC 1,800 mg/day after partial response to 600 mg/day, and then 1,200 mg/day.Citation210 A more recent report highlighted the use of NAC in 35 patients with Prader–Willi syndrome (PWS) and skin-picking behaviors. Patients diagnosed with PWS were aged 5–39 years (23 females/12 males) and treatment consisted of NAC at 450–1,200 mg/day for 12 weeks.Citation175 At treatment conclusion, all patients had reduced (n=10) or absent (n=25) skin-picking behaviors.Citation175

Lamotrigine

An open-label trial of lamotrigine 25–300 mg/day for the management of skin-picking behaviors showed that 67% of subjects were much or very much improved, prompting a randomized controlled trial.Citation211 A double-blind, placebo-controlled, randomized trial of lamotrigine for ExD showed a nonsignificant benefit of active drug (44% responders) compared to placebo (31% responders). Only those patients with difficulties in a test of shifting (cognitive flexibility) benefited maximally from the use of lamotrigine.Citation162

Topiramate

An open-label, 16-week trial of topiramate 50–250 mg/day in 14 adults with TTM resulted in nine adults completing the study.Citation212 Trial completers had a significant reduction of hair-pulling behaviors, with 6/9 classified as responders. Five patients dropped out due to adverse effects.Citation212 In a model of ExD using patients with PWS, topiramate 50–150 mg/day was administered as an add-on for the management of self-injury. All three patients experienced a significant resolution of ExD within 8 weeks of initiating the topiramate trial.Citation213 Another case reportCitation214 noted that two adolescents, aged 15 years and 16 years, with autism spectrum disorder and significant ExD symptoms benefited from topiramate addition to their psychotropic regimen in doses up to 200 mg/day for 12 weeks.

Riluzole

A 52-year-old female with OCD, anorexia, and major depression, who also exhibited ExD, was treated with fluoxetine, to which riluzole 100 mg bid was added. Scores on OCD rating, eating disordered behaviors, and pathologic skin picking all improved on this regimen. It was noted that urges and compulsive behaviors had diminished notably with the addition of riluzole.Citation215

Conclusion

CNS homeostasis of inhibition–disinhibition signaling ultimately depends on a well-regulated glutamate–GABA balance, in conjunction with other neurotransmitter systems that impact on this final effector pathway, which highly impacts neuronal health.Citation216 Future drug design approaches will benefit from a better understanding of these pathways (which also impact on other biologic systems, including immune and developmental networks) in OCD and related disorders, tics and ADHD, in order to provide a paradigmatic framework to better understand the imbalance in inhibition–disinhibition from the molecular level (glutamate–GABA) to the macrobehavioral level (obsessions, compulsions, tics, hyperactivity, and grooming behaviors). The heuristic value of considering these cross-disorder clinical manifestations in toto in relation to glutamate awaits future drug discovery to address these disinhibitory phenomena. In summary, drugs that impact the glutamatergic balance in the CNS are emerging as a therapeutic alternative for neuropsychiatric disorders, which implicate abnormal inhibitory control in cognitive, motor, behavioral, and grooming domains. OCD has the most support at present for the use of glutamate modulators, with ADHD and grooming disorders also showing promise. TS appears to be less affected by glutamate modulators, but the research is quite sparse in this condition. Future research into the pathophysiology of glutamate actions, and resultant drug discovery, in the CNS should include its role in early development,Citation217 neurogenesis,Citation218 and trophic mechanisms.Citation219

Disclosure

The authors report no conflicts of interest in this work.

References

  • MerrittKMcGuirePEgertonARelationship between Glutamate Dysfunction and Symptoms and Cognitive Function in PsychosisFront Psychiatry2013415124324444
  • HashimotoKMalchowBFalkaiPSchmittAGlutamate modulators as potential therapeutic drugs in schizophrenia and affective disordersEur Arch Psychiatry Clin Neurosci2013263536737723455590
  • MusazziLTreccaniGMalleiAPopoliMThe action of antidepressants on the glutamate system: regulation of glutamate release and glutamate receptorsBiol Psychiatry201373121180118823273725
  • CoyleJTBasuABenneyworthMBaluDKonopaskeGGlutamatergic synaptic dysregulation in schizophrenia: therapeutic implicationsHandb Exp Pharmacol201221326729523027419
  • American Psychiatric AssociationDiagnostic and Statistical Manual of Mental Disorders5th edWashington, DCAmerican Psychiatric Association2014
  • LeckmanJFGriceDEBoardmanJSymptoms of obsessive-compulsive disorderAm J Psychiatry199715479119179210740
  • RuscioAMSteinDJChiuWTKesslerRCThe epidemiology of obsessive-compulsive disorder in the National Comorbidity Survey ReplicationMol Psychiatry2010151536318725912
  • HeymanIFombonneESimmonsHFordTMeltzerHGoodmanRPrevalence of obsessive-compulsive disorder in the British nationwide survey of child mental healthBr J Psychiatry200117932432911581112
  • CraskeMGTreanorMConwayCCZbozinekTVervlietBMaximizing exposure therapy: an inhibitory learning approachBehav Res Ther201458102324864005
  • Pediatric OCD Treatment Study (POTS) TeamCognitive-behavior therapy, sertraline, and their combination for children and adolescents with obsessive-compulsive disorder: the Pediatric OCD Treatment Study (POTS) randomized controlled trialJAMA2004292161969197615507582
  • RosenbergDRKeshavanMSAE Bennett Research Award. Toward a neurodevelopmental model of obsessive – compulsive disorderBiol Psychiatry19984396236409582996
  • LujánRShigemotoRLópez-BenditoGGlutamate and GABA receptor signalling in the developing brainNeuroscience2005130356758015590141
  • MartinLJFurutaABlackstoneCDAMPA receptor protein in developing rat brain: glutamate receptor-1 expression and localization change at regional, cellular, and subcellular levels with maturationNeuroscience19988339179289483574
  • FurutaAMartinLJLaminar segregation of the cortical plate during corticogenesis is accompanied by changes in glutamate receptor expressionJ Neurobiol1999391678010213454
  • SattlerRTymianskiMMolecular mechanisms of glutamate receptor-mediated excitotoxic neuronal cell deathMol Neurobiol2001241–310712911831548
  • GradosMASpechtMWSungHMFortuneDGlutamate drugs and pharmacogenetics of OCD: a pathway-based exploratory approachExpert Opin Drug Discov20138121515152724147578
  • RosenbergDRMacMasterFPKeshavanMSFitzgeraldKDStewartCMMooreGJDecrease in caudate glutamatergic concentrations in pediatric obsessive-compulsive disorder patients taking paroxetineJ Am Acad Child Adolesc Psychiatry20003991096110310986805
  • MacMasterFPO’NeillJRosenbergDRBrain imaging in pediatric obsessive-compulsive disorderJ Am Acad Child Adolesc Psychiatry200847111262127218827717
  • ArnoldPDSicardTBurroughsERichterMAKennedyJLGlutamate transporter gene SLC1A1 associated with obsessive-compulsive disorderArch Gen Psychiatry200663776977616818866
  • StewartSEFagernessJAPlatkoJAssociation of the SLC1A1 glutamate transporter gene and obsessive-compulsive disorderAm J Med Genet B Neuropsychiatr Genet2007144B81027103317894418
  • DickelDEVeenstra-VanderWeeleJCoxNJAssociation testing of the positional and functional candidate gene SLC1A1/EAAC1 in early-onset obsessive-compulsive disorderArch Gen Psychiatry200663777878516818867
  • ShugartYYWangYSamuelsJFA family-based association study of the glutamate transporter gene SLC1A1 in obsessive-compulsive disorder in 378 familiesAm J Med Genet B Neuropsychiatr Genet2009150B688689219152386
  • StewartSEMayerfeldCArnoldPDMeta-analysis of association between obsessive-compulsive disorder and the 3′ region of neuronal glutamate transporter gene SLC1A1Am J Med Genet B Neuropsychiatr Genet2013162B436737923606572
  • ArnoldPDMacmasterFPRichterMAGlutamate receptor gene (GRIN2B) associated with reduced anterior cingulate glutamatergic concentration in pediatric obsessive-compulsive disorderPsychiatry Res2009172213613919324536
  • AlonsoPGratacósMSegalàsCAssociation between the NMDA glutamate receptor GRIN2B gene and obsessive-compulsive disorderJ Psychiatry Neurosci201237427328122433450
  • TükelROzataBOztürkNErtekinBAErtekinEDireskeneliGSThe role of the brain-derived neurotrophic factor SNP rs2883187 in the phenotypic expression of obsessive-compulsive disorderJ Clin Neurosci201421579079324291483
  • MárquezLCamarenaBHernándezSLóyzagaCVargasLNicoliniHAssociation study between BDNF gene variants and Mexican patients with obsessive-compulsive disorderEur Neuropsychopharmacol201323111600160523999029
  • TükelRGürvitHOzataBBrain-derived neurotrophic factor gene Val66Met polymorphism and cognitive function in obsessive-compulsive disorderAm J Med Genet B Neuropsychiatr Genet2012159B785085822911909
  • GreenbergBDZiemannUCorá-LocatelliGAltered cortical excitability in obsessive-compulsive disorderNeurology200054114214710636140
  • PittengerCKrystalJHCoricVGlutamate-modulating drugs as novel pharmacotherapeutic agents in the treatment of obsessive-compulsive disorderNeuroRx200631698116490414
  • NicolasLBKolbYPrinssenEPA combined marble burying-locomotor activity test in mice: a practical screening test with sensitivity to different classes of anxiolytics and antidepressantsEur J Pharmacol20065471–310611516934246
  • ShimazakiTIijimaMChakiSAnxiolytic-like activity of MGS0039, a potent group II metabotropic glutamate receptor antagonist, in a marble-burying behavior testEur J Pharmacol20045011–312112515464070
  • WelchJMLuJRodriguizRMCortico-striatal synaptic defects and OCD-like behaviours in Sapap3-mutant miceNature2007448715689490017713528
  • ChenMWanYAdeKTingJFengGCalakosNSapap3 deletion anomalously activates short-term endocannabinoid-mediated synaptic plasticityJ Neurosci201131269563957321715621
  • JoelDAvisarAExcessive lever pressing following post-training signal attenuation in rats: a possible animal model of obsessive compulsive disorder?Behav Brain Res20011231778711377731
  • AlbeldaNBar-OnNJoelDThe role of NMDA receptors in the signal attenuation rat model of obsessive-compulsive disorderPsychopharmacology (Berl)20102101132420238210
  • Alves GalvãoMGRochaCrispinoSantosMAAlves da CunhaAJAmantadine and rimantadine for influenza A in children and the elderlyCochrane Database Syst Rev20081CD00274518254006
  • WilliamsSEAmantadine treatment following traumatic brain injury in childrenBrain Inj200721988588917729042
  • DraytonSJDaviesKSteinbergMLeroiIRosenblattALyketsosCGAmantadine for executive dysfunction syndrome in patients with dementiaPsychosomatics200445320520915123844
  • HubsherGHaiderMOkunMSAmantadine: the journey from fighting flu to treating Parkinson diseaseNeurology201278141096109922474298
  • HosenbocusSChahalRAmantadine: a review of use in child and adolescent psychiatryJ Can Acad Child Adolesc Psychiatry2013221556023390434
  • NeagoeADDelirium with manic and psychotic features associated with amantadineGen Hosp Psychiatry2013356680.e7e823731892
  • PasquiniMBerardelliIBiondiMAmantadine augmentation for refractory obsessive-compulsive disorder: a case reportJ Clin Psychopharmacol2010301858620075660
  • StryjerRBudnikDEbertTAmantadine augmentation therapy for obsessive compulsive patients resistant to SSRIs-an open-label studyClin Neuropharmacol2014373798124824662
  • MyskiwJCIzquierdoIFuriniCRModulation of the extinction of fear learningBrain Res Bull2014105616924742526
  • NorbergMMKrystalJHTolinDFA meta-analysis of D-cycloserine and the facilitation of fear extinction and exposure therapyBiol Psychiatry200863121118112618313643
  • LeeHKimJJAmygdalar NMDA receptors are critical for new fear learning in previously fear-conditioned ratsJ Neurosci19981820844484549763487
  • HofmannSGPollackMHOttoMWAugmentation treatment of psychotherapy for anxiety disorders with D-cycloserineCNS Drug Rev2006123–420821717227287
  • KushnerMGKimSWDonahueCD-cycloserine augmented exposure therapy for obsessive-compulsive disorderBiol Psychiatry200762883583817588545
  • WilhelmSBuhlmannUTolinDFAugmentation of behavior therapy with D-cycloserine for obsessive-compulsive disorderAm J Psychiatry20081653335341 quiz 40918245177
  • StorchEAMerloLJBengtsonMD-cycloserine does not enhance exposure-response prevention therapy in obsessive-compulsive disorderInt Clin Psychopharmacol200722423023717519647
  • RodriguesHFigueiraILopesADoes D-cycloserine enhance exposure therapy for anxiety disorders in humans? A meta-analysisPLoS One201497e9351924991926
  • HernandesMSTronconeLRGlycine as a neurotransmitter in the forebrain: a short reviewJ Neural Transm2009116121551156019826900
  • ClementsJDWestbrookGLActivation kinetics reveal the number of glutamate and glycine binding sites on the N-methyl-D-aspartate receptorNeuron1991746056131681832
  • LabrieVRoderJCThe involvement of the NMDA receptor D-serine/glycine site in the pathophysiology and treatment of schizophreniaNeurosci Biobehav Rev201034335137219695284
  • GreenbergWMBenedictMMDoerferJAdjunctive glycine in the treatment of obsessive-compulsive disorder in adultsJ Psychiatr Res200943666467019046587
  • KrystalJHKarperLPSeibylJPSubanesthetic effects of the noncompetitive NMDA antagonist, ketamine, in humans. Psychotomimetic, perceptual, cognitive, and neuroendocrine responsesArch Gen Psychiatry19945131992148122957
  • BermanRMCappielloAAnandAAntidepressant effects of ketamine in depressed patientsBiol Psychiatry200047435135410686270
  • CaddyCGiaroliGWhiteTPShergillSSTracyDKKetamine as the prototype glutamatergic antidepressant: pharmacodynamic actions, and a systematic review and meta-analysis of efficacyTher Adv Psychopharmacol201442759924688759
  • RodriguezCIKegelesLSFloodPSimpsonHBRapid resolution of obsessions after an infusion of intravenous ketamine in a patient with treatment-resistant obsessive-compulsive disorderJ Clin Psychiatry201172456756921527129
  • BlochMHWasylinkSLanderos-WeisenbergerAEffects of ketamine in treatment-refractory obsessive-compulsive disorderBiol Psychiatry2012721196497022784486
  • RodriguezCIKegelesLSLevinsonARandomized controlled crossover trial of ketamine in obsessive-compulsive disorder: proof-of-conceptNeuropsychopharmacology201338122475248323783065
  • XieXHaganRMCellular and molecular actions of lamotrigine: Possible mechanisms of efficacy in bipolar disorderNeuropsychobiology19983831191309778599
  • CunninghamMOJonesRSThe anticonvulsant, lamotrigine decreases spontaneous glutamate release but increases spontaneous GABA release in the rat entorhinal cortex in vitroNeuropharmacology200039112139214610963757
  • BursteinAHLamotriginePharmacotherapy19951521291437624259
  • KetterTAManjiHKPostRMPotential mechanisms of action of lamotrigine in the treatment of bipolar disordersJ Clin Psychopharmacol200323548449514520126
  • BowdenCLCalabreseJRSachsGLamictal 606 Study GroupA placebo-controlled 18-month trial of lamotrigine and lithium maintenance treatment in recently manic or hypomanic patients with bipolar I disorderArch Gen Psychiatry200360439240012695317
  • CalabreseJRSuppesTBowdenCLA double-blind, placebo-controlled, prophylaxis study of lamotrigine in rapid-cycling bipolar disorder. Lamictal 614 Study GroupJ Clin Psychiatry2000611184185011105737
  • ThomasSPNandhraHSJayaramanASystematic review of lamotrigine augmentation of treatment resistant unipolar depression (TRD)J Ment Health201019216817520433324
  • HertzbergMAButterfieldMIFeldmanMEA preliminary study of lamotrigine for the treatment of posttraumatic stress disorderBiol Psychiatry19994591226122910331117
  • WaldmeierPCBaumannPAWickiPFeldtrauerJJStierlinCSchmutzMSimilar potency of carbamazepine, oxcarbazepine, and lamotrigine in inhibiting the release of glutamate and other neurotransmittersNeurology19954510190719137477991
  • UzunOLamotrigine as an augmentation agent in treatment-resistant obsessive-compulsive disorder: a case reportJ Psychopharmacol201024342542719010977
  • BrunoAMicòUPandolfoGLamotrigine augmentation of serotonin reuptake inhibitors in treatment-resistant obsessive-compulsive disorder: a double-blind, placebo-controlled studyJ Psychopharmacol201226111456146222351381
  • ErdöSLSchäferMMemantine is highly potent in protecting cortical cultures against excitotoxic cell death evoked by glutamate and N-methyl-D-aspartateEur J Pharmacol19911982–32152171864308
  • KornhuberJWellerMSchoppmeyerKRiedererPAmantadine and memantine are NMDA receptor antagonists with neuroprotective propertiesJ Neural Transm Suppl199443911047884411
  • EmreMTsolakiMBonuccelliU11018 Study InvestigatorsMemantine for patients with Parkinson’s disease dementia or dementia with Lewy bodies: a randomised, double-blind, placebo-controlled trialLancet Neurol201091096997720729148
  • CankurtaranESOzalpESoygurHCakirAClinical experience with risperidone and memantine in the treatment of Huntington’s diseaseJ Natl Med Assoc20069881353135516916137
  • TariotPNFarlowMRGrossbergGTGrahamSMMcDonaldSGergelIMemantine Study GroupMemantine treatment in patients with moderate to severe Alzheimer disease already receiving donepezil: a randomized controlled trialJAMA2004291331732414734594
  • HezelDMBeattieKStewartSEMemantine as an augmenting agent for severe pediatric OCDAm J Psychiatry2009166223719188297
  • PasquiniMBiondiMMemantine augmentation for refractory obsessive-compulsive disorderProg Neuropsychopharmacol Biol Psychiatry20063061173117516730870
  • AboujaoudeEBarryJJGamelNMemantine augmentation in treatment-resistant obsessive-compulsive disorder: an open-label trialJ Clin Psychopharmacol2009291515519142108
  • FeusnerJDKerwinLSaxenaSBystritskyADifferential efficacy of memantine for obsessive-compulsive disorder vs generalized anxiety disorder: an open-label trialPsychopharmacol Bull2009421819319204653
  • GhaleihaAEntezariNModabberniaAMemantine add-on in moderate to severe obsessive-compulsive disorder: randomized double-blind placebo-controlled studyJ Psychiatr Res201347217518023063327
  • BlumsteinCGDrug treatment in bronchial asthmaSemin Drug Treat1973243854014581867
  • SteyCSteurerJBachmannSMediciTCTramèrMRThe effect of oral N-acetylcysteine in chronic bronchitis: a quantitative systematic reviewEur Respir J200016225326210968500
  • SadowskaAMVerbraeckenJDarquennesKDe BackerWARole of N-acetylcysteine in the management of COPDInt J Chron Obstruct Pulmon Dis20061442543418044098
  • RumackBHAcetaminophen overdose in children and adolescentsPediatr Clin North Am19863336917013714342
  • KearnsGLLeederJSWassermanGSAcetaminophen intoxication during treatment: what you don’t know can hurt youClin Pediatr (Phila)200039313314410752006
  • HeardKGreenJAcetylcysteine therapy for acetaminophen poisoningCurr Pharm Biotechnol201213101917192322352734
  • AricaVDemirİHTutancMN-acetylcysteine prevents doxorubucine-induced cardiotoxicity in ratsHum Exp Toxicol201332665566123424206
  • Soltan-SharifiMSMojtahedzadehMNajafiAImprovement by N-acetylcysteine of acute respiratory distress syndrome through increasing intracellular glutathione, and extracellular thiol molecules and anti-oxidant power: evidence for underlying toxicological mechanismsHum Exp Toxicol200726969770317984140
  • WuYJMuldoonLLNeuweltEAThe chemoprotective agent N-acetylcysteine blocks cisplatin-induced apoptosis through caspase signaling pathwayJ Pharmacol Exp Ther2005312242443115496615
  • OrhanGYapiciNYukselMEffects of N-acetylcysteine on myocardial ischemia-reperfusion injury in bypass surgeryHeart Vessels2006211424716440148
  • Borges-SantosMDMoretoFPereiraPCMing-YuYBuriniRCPlasma glutathione of HIV+ patients responded positively and differently to dietary supplementation with cysteine or glutamineNutrition2012287–875375622261571
  • KandemirHAbuhandanMAksoyNSavikEKayaCOxidative imbalance in child and adolescent patients with obsessive compulsive disorderJ Psychiatr Res201347111831183424011862
  • ErsanSBakirSErdal ErsanEDoganOExamination of free radical metabolism and antioxidant defence system elements in patients with obsessive-compulsive disorderProg Neuropsychopharmacol Biol Psychiatry20063061039104216682105
  • LafleurDLPittengerCKelmendiBN-acetylcysteine augmentation in serotonin reuptake inhibitor refractory obsessive-compulsive disorderPsychopharmacology (Berl)2006184225425616374600
  • WikkeJRiluzoleLancet199634890307957998813989
  • JehleTBauerJBlauthEEffects of riluzole on electrically evoked neurotransmitter releaseBr J Pharmacol200013061227123410903959
  • UrbaniABelluzziORiluzole inhibits the persistent sodium current in mammalian CNS neuronsEur J Neurosci200012103567357411029626
  • SreedharanJBrownRHJrAmyotrophic lateral sclerosis: problems and prospectsAnn Neurol201374330931624038380
  • ClarkWKendallMJTherapeutic advances: riluzole for the treatment of motor neurone diseaseJ Clin Pharm Ther19962163733769201563
  • GordonPHAmyotrophic lateral sclerosis: an update for 2013 clinical features, pathophysiology, management and therapeutic trialsAging Dis20134529531024124634
  • LacomblezLBensimonGLeighPNALS Study Groups I and IILong-term safety of riluzole in amyotrophic lateral sclerosisAmyotroph Lateral Scler Other Motor Neuron Disord200231232912061945
  • CoricVTaskiranSPittengerCRiluzole augmentation in treatment-resistant obsessive-compulsive disorder: an open-label trialBiol Psychiatry200558542442815993857
  • GrantPLougeeLHirschtrittMSwedoSEAn open-label trial of riluzole, a glutamate antagonist, in children with treatment-resistant obsessive-compulsive disorderJ Child Adolesc Psychopharmacol200717676176718315448
  • GrantPJJosephLAFarmerCA12-week, placebo-controlled trial of add-on riluzole in the treatment of childhood-onset obsessive-compulsive disorderNeuropsychopharmacology20143961453145924356715
  • GarnettWRClinical pharmacology of topiramate: a reviewEpilepsia200041Suppl 1S61S6510768303
  • BauerJSchwalenSTopiramate (Topamax). Pharmacological characteristics and current use in epilepsy treatmentNervenarzt2000716495501 German10919147
  • PoulsenCFSimeoneTAMaarTESmith-SwintoskyVWhiteHSSchousboeAModulation by topiramate of AMPA and kainate mediated calcium influx in cultured cerebral cortical, hippocampal and cerebellar neuronsNeurochem Res200429127528214992287
  • ManningSPucciAFinerNPharmacotherapy for obesity: novel agents and paradigmsTher Adv Chronic Dis20145313514824790728
  • LindeMMullenersWMChronicleEPMcCroryDCTopiramate for the prophylaxis of episodic migraine in adultsCochrane Database Syst Rev20136CD01061023797676
  • HollanderEDell’OssoBTopiramate plus paroxetine in treatment-resistant obsessive-compulsive disorderInt Clin Psychopharmacol200621318919116528143
  • Van AmeringenMManciniCPattersonBBennettMTopiramate augmentation in treatment-resistant obsessive-compulsive disorder: a retrospective, open-label case seriesDepress Anxiety20062311516178009
  • BerlinHAKoranLMJenikeMADouble-blind, placebo- controlled trial of topiramate augmentation in treatment-resistant obsessive-compulsive disorderJ Clin Psychiatry201172571672120816027
  • OzkaraCOzmenMErdoganAYalugITopiramate related obsessive-compulsive disorderEur Psychiatry2005201787915642451
  • Lauria-HornerBAPohlRBPregabalin: a new anxiolyticExpert Opin Investig Drugs2003124663672
  • MichevaKDTaylorCPSmithSJPregabalin reduces the release of synaptic vesicles from cultured hippocampal neuronsMol Pharmacol200670246747616641316
  • OulisPMourikisIKonstantakopoulosGPregabalin augmentation in treatment-resistant obsessive-compulsive disorderInt Clin Psychopharmacol201126422122421460732
  • RampelloLAlvanoABattagliaGBrunoVRaffaeleRNicolettiFTic disorders: from pathophysiology to treatmentJ Neurol2006253111516331353
  • SpechtMWNicotraCMKellyLMA Comparison of urge intensity and the probability of tic completion during tic freely and tic suppression conditionsBehav Modif201438229731824924158
  • WoodsDWPiacentiniJCScahillLBehavior therapy for tics in children: acute and long-term effects on psychiatric and psychosocial functioningJ Child Neurol201126785886521555779
  • WilhelmSPetersonALPiacentiniJRandomized trial of behavior therapy for adults with Tourette syndromeArch Gen Psychiatry201269879580322868933
  • RizzoRGulisanoMClinical pharmacology of comorbid attention deficit hyperactivity disorder in Tourette syndromeInt Rev Neurobiol201311241544424295629
  • NeriVCardonaFClinical pharmacology of comorbid obsessive-compulsive disorder in Tourette syndromeInt Rev Neurobiol201311239141424295628
  • RizzoRGulisanoMPellicoACalìPVCuratoloPTourette syndrome and comorbid conditions: a spectrum of different severities and complexitiesJ Child Neurol201429101383138924832397
  • BerardelliACurràAFabbriniGGilioFManfrediMPathophysiology of tics and Tourette syndromeJ Neurol2003250778178712883917
  • SingerHSMinzerKNeurobiology of Tourette’s syndrome: concepts of neuroanatomic localization and neurochemical abnormalitiesBrain Dev200325Suppl 1S70S8414980376
  • NomuraYSegawaMNeurology of Tourette’s syndrome (TS) TS as a developmental dopamine disorder: a hypothesisBrain Dev200325Suppl 1S37S4214980371
  • YoonDYGauseCDLeckmanJFSingerHSFrontal dopaminergic abnormality in Tourette syndrome: a postmortem analysisJ Neurol Sci20072551–2505617337006
  • UdvardiPTNespoliERizzoFHengererBLudolphAGNondopaminergic neurotransmission in the pathophysiology of Tourette syndromeInt Rev Neurobiol20131129513024295619
  • SingerHSMorrisCGradosMGlutamatergic modulatory therapy for Tourette syndromeMed Hypotheses201074586286720022434
  • AndersonGMPollakESChatterjeeDLeckmanJFRiddleMACohenDJPostmortem analysis of subcortical monoamines and amino acids in Tourette syndromeAdv Neurol1992581231331414615
  • SwerdlowNRSutherlandANUsing animal models to develop therapeutics for Tourette SyndromePharmacol Ther2005108328129315970330
  • McGrathMJCampbellKMParksCRBurtonFHGlutamatergic drugs exacerbate symptomatic behavior in a transgenic model of comorbid Tourette’s syndrome and obsessive-compulsive disorderBrain Res20008771233010980239
  • NordstromEJBurtonFHA transgenic model of comorbid Tourette’s syndrome and obsessive-compulsive disorder circuitryMol Psychiatry20027661762552412140785
  • ShoemakerJMSaint MarieRLBongiovanniMJNearyACTochenLSSwerdlowNRPrefrontal D1 and ventral hippocampal N-methyl-D-aspartate regulation of startle gating in ratsNeuroscience2005135238539416125865
  • AbelKMAllinMPHemsleyDRGeyerMALow dose ketamine increases prepulse inhibition in healthy menNeuropharmacology200344672973712681371
  • DuncanEJMadonickSHParwaniAClinical and sensorimotor gating effects of ketamine in normalsNeuropsychopharmacology2001251728311377920
  • OrtizAAldaMO’DonovanCTourette syndrome with comorbid bipolar disorder and migraine: can lamotrigine monotherapy help?J Clin Psychopharmacol201232114014222217952
  • SeemüllerFDehningSGrunzeHMüllerNTourette’s symptoms provoked by lamotrigine in a bipolar patientAm J Psychiatry2006163115916390908
  • LombrosoCTLamotrigine-induced tourettismNeurology19995261191119410214742
  • Sotero de MenezesMARhoJMMurphyPCheyetteSLamotrigine-induced tic disorder: report of five pediatric casesEpilepsia200041786286710897158
  • RobertsonMMEapenVCavannaAEThe international prevalence, epidemiology, and clinical phenomenology of Tourette syndrome: a cross-cultural perspectiveJ Psychosom Res200967647548319913651
  • FornaroMMaremmaniAGColicchioMGA case of severe oral self-injurious Tourette’s syndrome alleviated by pregabalinGen Hosp Psychiatry2012343321.e1e422133981
  • KuoSHJimenez-ShahedJTopiramate in treatment of tourette syndromeClin Neuropharmacol2010331323419935407
  • JankovicJJimenez-ShahedJBrownLWA randomised, double-blind, placebo-controlled study of topiramate in the treatment of Tourette syndromeJ Neurol Neurosurg Psychiatry2010811707319726418
  • UnderhillSMWheelerDSLiMWattsSDIngramSLAmaraSGAmphetamine modulates excitatory neurotransmission through endocytosis of the glutamate transporter EAAT3 in dopamine neuronsNeuron201483240441625033183
  • RollandBJardriRAmadAThomasPCottencinOBordetRPharmacology of hallucinations: several mechanisms for one single symptom?Biomed Res Int2014201430710624991548
  • TaylorMBhagwandasKN-acetylcysteine in trichotillomania: a panacea for compulsive skin disorders?Br J Dermatol201417151253125524773206
  • Van AmeringenMPattersonBSimpsonWDSM-5 obsessive-compulsive and related disorders: clinical implications of new criteriaDepress Anxiety201431648749324616177
  • MasurDShinnarSCnaanAChildhood Absence Epilepsy Study GroupPretreatment cognitive deficits and treatment effects on attention in childhood absence epilepsyNeurology201381181572158024089388
  • MalloMAlonsoCRThe regulation of Hox gene expression during animal developmentDevelopment2013140193951396324046316
  • PanzaKEPittengerCBlochMHAge and gender correlates of pulling in pediatric trichotillomaniaJ Am Acad Child Adolesc Psychiatry201352324124923452681
  • BlochMHPanzaKEGrantJEPittengerCLeckmanJFN-Acetylcysteine in the treatment of pediatric trichotillomania: a randomized, double-blind, placebo-controlled add-on trialJ Am Acad Child Adolesc Psychiatry201352323124023452680
  • Rodrigues-BarataARTostiARodríguez-PichardoACamacho-MartínezFN-acetylcysteine in the treatment of trichotillomaniaInt J Trichology20124317617823180931
  • GrantJEOdlaugBLChamberlainSRKeuthenNJLochnerCSteinDJSkin picking disorderAm J Psychiatry2012169111143114923128921
  • Schneebaum-SenderNGoldberg-SternHFattal-ValevskiAKramerUDoes a normalizing electroencephalogram in benign childhood epilepsy with centrotemporal spikes abort attention deficit hyperactivity disorder?Pediatr Neurol201247427928322964442
  • WuJXiaoHSunHZouLZhuLQRole of dopamine receptors in ADHD: a systematic meta-analysisMol Neurobiol201245360562022610946
  • GrantJEOdlaugBLChamberlainSRA cognitive comparison of pathological skin picking and trichotillomaniaJ Psychiatr Res201145121634163821824627
  • ChamberlainSRHampshireAMenziesLAReduced brain white matter integrity in trichotillomania: a diffusion tensor imaging studyArch Gen Psychiatry201067996597120819990
  • GrantJEOdlaugBLChamberlainSRKimSWA double-blind, placebo-controlled trial of lamotrigine for pathological skin picking: treatment efficacy and neurocognitive predictors of responseJ Clin Psychopharmacol201030439640320531220
  • GrantJEOdlaugBLKimSWN-acetylcysteine, a glutamate modulator, in the treatment of trichotillomania: a double-blind, placebo-controlled studyArch Gen Psychiatry200966775676319581567
  • ChamberlainSROdlaugBLBoulougourisVFinebergNAGrantJETrichotillomania: neurobiology and treatmentNeurosci Biobehav Rev200933683184219428495
  • PaineTANeveRLCarlezonWAJrAttention deficits and hyperactivity following inhibition of cAMP-dependent protein kinase within the medial prefrontal cortex of ratsNeuropsychopharmacology20093492143215519387423
  • TsengKYO’DonnellPDopamine-glutamate interactions controlling prefrontal cortical pyramidal cell excitability involve multiple signaling mechanismsJ Neurosci200424225131513915175382
  • MacMasterFPCarreyNSparkesSKusumakarVProton spectroscopy in medication-free pediatric attention-deficit/hyperactivity disorderBiol Psychiatry200353218418712547476
  • CarreyNMacMasterFPFogelJMetabolite changes resulting from treatment in children with ADHD: a 1H-MRS studyClin Neuropharmacol200226421822112897644
  • CarreyNMacMasterFPSparkesSJKhanSCKusumakarVGlutamatergic changes with treatment in attention deficit hyperactivity disorder: a preliminary case seriesJ Child Adolesc Psychopharmacol200212433133612625993
  • BushGLuuPPosnerMICognitive and emotional influences in anterior cingulate cortexTrends Cogn Sci20004621522210827444
  • HammernessPBiedermanJPettyCHeninAMooreCMBrain biochemical effects of methylphenidate treatment using proton magnetic spectroscopy in youth with attention-deficit hyperactivity disorder: a controlled pilot studyCNS Neurosci Ther2012181344021143432
  • XuMMoratallaRGoldLHDopamine D1 receptor mutant mice are deficient in striatal expression of dynorphin and in dopamine-mediated behavioral responsesCell19947947297427954836
  • GranonSPassettiFThomasKLDalleyJWEverittBJRobbinsTWEnhanced and impaired attentional performance after infusion of D1 dopaminergic receptor agents into rat prefrontal cortexJ Neurosci20002031208121510648725
  • WartonFLHowellsFMRussellVAIncreased glutamate-stimulated release of dopamine in substantia nigra of a rat model for attention-deficit/hyperactivity disorder – lack of effect of methylphenidateMetab Brain Dis200924459961319821016
  • MillerJLAnguloMAn open-label pilot study of N-acetylcysteine for skin-picking in Prader-Willi syndromeAm J Med Genet A2014164A242142424311388
  • LeschKPMerkerSReifANovakMDances with black widow spiders: dysregulation of glutamate signalling enters centre stage in ADHDEur Neuropsychopharmacol201323647949122939004
  • EliaJGlessnerJTWangKGenome-wide copy number variation study associates metabotropic glutamate receptor gene networks with attention deficit hyperactivity disorderNat Genet2012441788422138692
  • RubinsteinMCepedaCHurstRSDopamine D4 receptor-deficient mice display cortical hyperexcitabilityJ Neurosci200121113756376311356863
  • WangXZhongPGuZYanZRegulation of NMDA receptors by dopamine D4 signaling in prefrontal cortexJ Neurosci200323309852986114586014
  • GanLFalzoneTLZhangKRubinsteinMBaldessariniRJTaraziFIEnhanced expression of dopamine D(1) and glutamate NMDA receptors in dopamine D(4) receptor knockout miceJ Mol Neurosci200422316717814997010
  • ThomasTCGrandyDKGerhardtGAGlaserPEDecreased dopamine D4 receptor expression increases extracellular glutamate and alters its regulation in mouse striatumNeuropsychopharmacology200934243644518536704
  • ShaywitzAJGreenbergMECREB: a stimulus-induced transcription factor activated by a diverse array of extracellular signalsAnnu Rev Biochem19996882186110872467
  • PozziLBavieraMSacchettiGAttention 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 2/3Neuroscience201117633634821193020
  • ViggianoDValloneDWelzlHSadileAGThe Naples High- and Low-Excitability rats: selective breeding, behavioral profile, morphometry, and molecular biology of the mesocortical dopamine systemBehav Genet200232531533312405514
  • RuoccoLAGironi CarnevaleUASadileAGElevated forebrain excitatory L-glutamate, L-aspartate and D-aspartate in the Naples high-excitability ratsBehav Brain Res20091981242819073221
  • MattesJA pilot trial of amantadine in hyperactive childrenPsychopharmacol Bull198016367697403410
  • DonfrancescoRCalderoniDVitielloBOpen-label amantadine in children with attention-deficit/hyperactivity disorderJ Child Adolesc Psychopharmacol200717565766417979585
  • MohammadiMRKazemiMRZiaERezazadehSATabriziMAkhondzadehSAmantadine versus methylphenidate in children and adolescents with attention deficit/hyperactivity disorder: a randomized, double-blind trialHum Psychopharmacol2010257–856056521312290
  • ÖncüBErOÇolakBNuttDJLamotrigine for attention deficit-hyperactivity disorder comorbid with mood disorders: a case seriesJ Psychopharmacol201428328228323784736
  • UvebrantPBauzienèRIntractable epilepsy in children. The efficacy of lamotrigine treatment, including non-seizure-related benefitsNeuropediatrics19942562842897770124
  • SchubertRAttention deficit disorder and epilepsyPediatr Neurol200532111015607597
  • PeetersMMaloteauxJMHermansEDistinct effects of amantadine and memantine on dopaminergic transmission in the rat striatumNeurosci Lett2003343320520912770697
  • FindlingRLMcNamaraNKStansbreyRJA pilot evaluation of the safety, tolerability, pharmacokinetics, and effectiveness of memantine in pediatric patients with attention-deficit/hyperactivity disorder combined typeJ Child Adolesc Psychopharmacol2007171193317343551
  • SurmanCBHammernessPGPettyCA pilot open label prospective study of memantine monotherapy in adults with ADHDWorld J Biol Psychiatry201314429129822436083
  • GarciaRJFrancisLDawoodMLaiZWFaraoneSVPerlAAttention deficit and hyperactivity disorder scores are elevated and respond to N-acetylcysteine treatment in patients with systemic lupus erythematosusArthritis Rheum20136551313131823400548
  • ArnoldLMAuchenbachMBMcElroySLPsychogenic excoriation. Clinical features, proposed diagnostic criteria, epidemiology and approaches to treatmentCNS Drugs200115535135911475941
  • KeuthenNJKoranLMAboujaoudeELargeMDSerpeRTThe prevalence of pathologic skin picking in US adultsCompr Psychiatry201051218318620152300
  • HayesSLStorchEABerlangaLSkin picking behaviors: An examination of the prevalence and severity in a community sampleJ Anxiety Disord200923331431919223150
  • SwedoSERapoportJLAnnotation: trichotillomaniaJ Child Psychol Psychiatry19913234014092061361
  • NezirogluFRabinowitzDBreytmanAJacofskyMSkin picking phenomenology and severity comparisonPrim Care Companion J Clin Psychiatry200810430631218787665
  • BohneASavageCRDeckersbachTKeuthenNJWilhelmSMotor inhibition in trichotillomania and obsessive-compulsive disorderJ Psychiatr Res200842214115017215004
  • SwedoSERapoportJLLeonardHLSchapiroMBRapoportSIGradyCLRegional cerebral glucose metabolism of women with trichotillomaniaArch Gen Psychiatry19914898288331929773
  • RoosAFoucheJPSteinDJLochnerCWhite matter integrity in hair-pulling disorder (trichotillomania)Psychiatry Res2013211324625023149033
  • GrantJEOdlaugBLHampshireASchreiberLRChamberlainSRWhite matter abnormalities in skin picking disorder: a diffusion tensor imaging studyNeuropsychopharmacology201338576376923303052
  • StratfordTHKostakopoulouKMadenMHoxb-8 has a role in establishing early anterior-posterior polarity in chick forelimb but not hindlimbDevelopment199712421422542349334271
  • GreerJMCapecchiMRHoxb8 is required for normal grooming behavior in miceNeuron2002331233411779477
  • WelchJMWangDFengGDifferential mRNA expression and protein localization of the SAP90/PSD-95-associated proteins (SAPAPs) in the nervous system of the mouseJ Comp Neurol20044721243915024750
  • BienvenuOJWangYShugartYYSapap3 and pathological grooming in humans: Results from the OCD collaborative genetics studyAm J Med Genet B Neuropsychiatr Genet2009150B571072019051237
  • ZüchnerSWendlandJRAshley-KochAEMultiple rare SAPAP3 missense variants in trichotillomania and OCDMol Psychiatry20091416919096451
  • OdlaugBLGrantJEN-acetyl cysteine in the treatment of grooming disordersJ Clin Psychopharmacol200727222722917414258
  • GrantJEOdlaugBLKimSWLamotrigine treatment of pathologic skin picking: an open-label studyJ Clin Psychiatry20076891384139117915977
  • LochnerCSeedatSNiehausDJSteinDJTopiramate in the treatment of trichotillomania: an open-label pilot studyInt Clin Psychopharmacol200621525525916877895
  • ShapiraNALessigMCMurphyTKDriscollDJGoodmanWKTopiramate attenuates self-injurious behaviour in Prader-Willi SyndromeInt J Neuropsychopharmacol20025214114512135538
  • JafferanyMShireenFIbrahimAAn open-label trial of topiramate in the treatment of skin picking in pervasive developmental disorder not otherwise specifiedPrim Care Companion J Clin Psychiatry2010122piiPCC.09l00829
  • CoricVKelmendiBPittengerCWasylinkSBlochMHGreenJBeneficial effects of the antiglutamatergic agent riluzole in a patient diagnosed with trichotillomaniaJ Clin Psychiatry200768117017117284151
  • RothmanSMMattsonMPActivity-dependent, stress-responsive BDNF signaling and the quest for optimal brain health and resilience throughout the lifespanNeuroscience201323922824023079624
  • TanakaKBrain development and glutamateBrain Nerve2013651011211132 Japanese24101424
  • NakamichiNTakaradaTYonedaYNeurogenesis mediated by gamma-aminobutyric acid and glutamate signalingJ Pharmacol Sci2009110213314919483378
  • MattsonMPGlutamate and neurotrophic factors in neuronal plasticity and diseaseAnn N Y Acad Sci200811449711219076369