Next Article in Journal
Metformin Prevents NDEA-Induced Memory Impairments Associated with Attenuating Beta-Amyloid, Tumor Necrosis Factor-Alpha, and Interleukin-6 Levels in the Hippocampus of Rats
Next Article in Special Issue
Inhibition of NMDA Receptor Activation in the Rostral Ventrolateral Medulla by Amyloid-β Peptide in Rats
Previous Article in Journal
Metyrapone Treatment Protects Low-Density Lipoprotein Receptor Knockout Mice against Hypercorticosteronemia Development without Changing Atherosclerosis Susceptibility
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Novel Gliotransmitter, L-β-Aminoisobutyric Acid, Contributes to Pathophysiology of Clinical Efficacies and Adverse Reactions of Clozapine

Department of Neuropsychiatry, Division of Neuroscience, Graduate School of Medicine, Mie University, Tsu 514-8507, Japan
*
Author to whom correspondence should be addressed.
Biomolecules 2023, 13(9), 1288; https://doi.org/10.3390/biom13091288
Submission received: 28 July 2023 / Revised: 18 August 2023 / Accepted: 22 August 2023 / Published: 23 August 2023
(This article belongs to the Special Issue NMDA Receptor in Health and Diseases 2.0)

Abstract

:
Clozapine is listed as one of the most effective antipsychotics and has been approved for treating treatment-resistant schizophrenia (TRS); however, several type A and B adverse reactions, including weight gain, metabolic complications, cardiotoxicity, convulsions, and discontinuation syndromes, exist. The critical mechanisms of clinical efficacy for schizophrenia, TRS, and adverse reactions of clozapine have not been elucidated. Recently, the GABA isomer L-β-aminoisobutyric acid (L-BAIBA), a protective myokine in the peripheral organs, was identified as a candidate novel transmission modulator in the central nervous system (CNS). L-BAIBA activates adenosine monophosphate-activated protein kinase (AMPK) signalling in both the peripheral organs and CNS. Activated AMPK signalling in peripheral organs is an established major target for treating insulin-resistant diabetes, whereas activated AMPK signalling in the hypothalamus contributes to the pathophysiology of weight gain and metabolic disturbances. Clozapine increases L-BAIBA synthesis in the hypothalamus. In addition, the various functions of L-BAIBA in the CNS have recently been elucidated, including as an activator of GABA-B and group-III metabotropic glutamate (III-mGlu) receptors. Considering the expressions of GABA-B and III-mGlu receptors (localised in the presynaptic regions), the activation of GABA-B and III-mGlu receptors can explain the distinct therapeutic advantages of clozapine in schizophrenia or TRS associated with N-methyl-D-aspartate (NMDA) receptor disturbance compared with other atypical antipsychotics via the inhibition of the persistent tonic hyperactivation of thalamocortical glutamatergic transmission in the prefrontal cortex. L-BAIBA has also been identified as a gliotransmitter, and a detailed exploration of the function of L-BAIBA in tripartite synaptic transmission can further elucidate the pathophysiology of effectiveness for treating TRS and/or specific adverse reactions of clozapine.

1. Introduction

Traditionally, more than 30% of patients with schizophrenia spectrum are considered to suffer from treatment-resistant schizophrenia (TRS) [1,2,3]. Clozapine is evaluated as the most effective antipsychotic agent for TRS since 30–60% of patients with TRS respond to clozapine medication [4,5,6]. Therefore, clozapine is currently the only approved antipsychotic for TRS treatment [7]. In fact, several guidelines recommend initiating treatment with clozapine for patients with TRS [8,9,10]. Furthermore, systematic reviews and meta-analyses have demonstrated that clozapine is associated with lower hospitalisation rates, lower overall discontinuation rates, and better overall symptom outcomes compared with other atypical antipsychotics [11,12,13].
All antipsychotics approved for the treatment of schizophrenia are antagonists of the dopamine D2 receptor at therapeutically relevant concentrations [14,15]. The introduction of clozapine in the 1970s marked a significant turning point in the pharmacotherapy of schizophrenia. As an alternative, clozapine minimised the risk of extrapyramidal symptoms, such as antipsychotic-induced parkinsonism and tardive dyskinesia, while demonstrating excellent efficacy for both positive and negative symptoms of schizophrenia [16,17]. Based on these clinical advantages of clozapine, receptor-binding profile screenings have contributed to the development of several second-generation antipsychotics (atypical antipsychotics) that share pharmacological characteristics distinct from the preceding first-generation antipsychotics (typical antipsychotics) (Table 1) [16,18]. It is well known that olanzapine has a similar receptor-binding profile to clozapine, except for the 5-HT7 receptor [19] (Table 1); however, the specific effectiveness of clozapine for treating TRS suggests the pathophysiology of clozapine may involve molecules other than monoamine receptors.
Most atypical antipsychotics had been developed by exploring molecules that have similar receptor-binding profiles to clozapine that are distinct from the preceding typical antipsychotics, such as having a relatively lower binding affinity to the dopamine D2 receptor and higher affinity to serotonin 5-HT2A receptors [20]. Therefore, the pathophysiological hypothesis proposed to distinguish between typical and atypical antipsychotics, having a relatively low affinity to the D2 receptor and relatively high affinity to the 5-HT2A receptor, cannot account for the distinct therapeutic advantages of clozapine against other atypical antipsychotics.
Table 1. Receptor-binding profiles of antipsychotics.
Table 1. Receptor-binding profiles of antipsychotics.
ReceptorCLZLURAPZBrexOLZQTPRISZTPHPD
5-HT1A1246.85.60.12>1000432423471>1000
5-HT2A5.42.08.70.472.31000.22.753
5-HT2C9.4415766314>1000122.6>1000
5-HT718.00.510.33.73653076.612.0377
H11.13>100027.6191.21120.13.21>1000
D1266262>100016010071224471.080
D21571.73.30.352.32453.625.00.7
References[21,22][23][24,25][26][27,28][29][25,30][31][32,33]
Clozapine (CLZ), lurasidone (LUR), aripiprazole (APZ), brexpiprazole (Brex), olanzapine (OLZ), quetiapine (QTP), risperidone (RIS), zotepine (ZTP), and haloperidol (HPD) against serotonin (5-HT) type 1A (5-HT1A), type 2A (5-HT2A), type 2C (5-HT2C), and type 7 (5-HT7) receptors, histamine H1 (H1) receptor, and dopamine receptors type 1 (D1) and 2 (D2). Data are equilibrium constant (Ki) values (nM).
Despite the clinical advantages of clozapine’s effectiveness for TRS, clozapine is also associated with numerous specific/serious adverse reactions, such as type B reactions (agranulocytosis, eosinophilia and haematological malignancies, myocarditis, cardiomyopathy, and convulsions) and type A reactions (weight gain and metabolic disturbance) [34,35,36,37,38]. Occasionally, psychiatrists must promptly discontinue clozapine or switch to other antipsychotics due to these lethal type B adverse reactions. However, prompt discontinuation often leads to clozapine discontinuation symptoms, including clozapine-discontinuation-induced worsening of psychosis and catatonia [38,39,40]. These distinct clinical advantages and disadvantages of clozapine compared with conventional atypical antipsychotics suggest that clozapine likely has different mechanisms of action compared with other atypical antipsychotics.
Tripartite synaptic transmission involving molecules other than monoamine receptors has recently been speculated to play important roles in the pharmacological mechanisms of clozapine’s therapeutic effects and adverse reactions, which is distinct from other atypical antipsychotics [38,41,42,43,44,45,46,47]. In these pharmacodynamic researches, it has been identified that L-β-aminoisobutyric acid (L-BAIBA) plays an important role in the pathophysiology underlying the mechanisms of clozapine’s clinical efficacy for the treatment of schizophrenia and TRS as well as in adverse reactions, such as weight gain and metabolic complications [46,47]. In other words, L-BAIBA is a candidate target molecule that can rationally explain the mechanisms of clozapine’s therapeutic effects and adverse reactions, which have not been elucidated so far, via modulating various signalling pathways. In this review, we outline the characteristics of the therapeutic effects and adverse reactions of clozapine and then propose a compelling pathophysiological hypothesis that L-BAIBA is involved in the underlying mechanism of clozapine, which has not been elucidated.

2. Clozapine-Induced Metabolic Complications

Weight gain is the most prevalent adverse reaction of atypical antipsychotic medications. Weight gain induced by atypical antipsychotics usually occurs during the early stages of antipsychotic treatment (within the first year), with an increase of 7% over baseline weight observed in approximately two-thirds of antipsychotic-treated patients [48,49]. Diabetes treatment in patients treated with clozapine is manageable by following current diabetes treatment guidelines [50,51]. Thus, a history of diabetes in TRS patients does not constitute a contraindication to clozapine medication [52]. Among pharmacological interventions, metformin has an excellent safety profile and is the most effective for weight gain stabilisation [53,54,55]. Topiramate has also been demonstrated to be as effective as metformin in suppressive effects on clozapine-induced weight gain [56,57]. Glucagon-like peptide-1 (GLP1) receptor agonists have been recently shown to effectively mitigate clozapine-induced metabolic disturbances [58]. However, weight gain induced by antipsychotics other than clozapine, including olanzapine and quetiapine, reaches a plateau within the therapeutic dose range, whereas the unique features of weight gain with clozapine indicate a linear dose-dependent manner ranging from therapeutic to supratherapeutic doses [59]. This specific linear dose-dependent weight gain induced by clozapine indicates that different mechanisms might underlie the weight gain induced by other antipsychotics.
Atypical antipsychotic-induced metabolic complications have been considered to be related to the inhibition of the histamine H1 and serotonin 5-HT2A receptors, which leads to the disturbance of energy regulation systems in the hypothalamus [60,61]. The inhibition of the H1 and 5-HT2A receptors suppresses the synthesis of inositol trisphosphate (IP3), which activates the calcium-induced calcium-releasing system (CICR) via the enhancement of the IP3 receptor (Figure 1) [62,63]. The elevation in intracellular calcium ion levels activates adenosine triphosphate (ATP) synthase, leading to an increase in ATP and/or a decrease in adenosine monophosphate (AMP) levels (Figure 1) [61,63,64]. Therefore, CICR suppression induced by H1 and 5-HT2A receptor inhibition secondarily increases intracellular AMP levels, leading to the activation of adenosine monophosphate (AMP)-activated protein kinase (AMPK) (Figure 1) [46,60,61,64]. This hypothesis has been supported by the clinical findings on high-affinity H1 and 5-HT2A receptor antagonistic antipsychotics, including zotepine, quetiapine, olanzapine, and clozapine listed as being high-risk for metabolic complications [59]. However, the activation of AMPK in the peripheral organs is one of the major therapeutic targets for insulin-resistant diabetes [55,65,66], whereas the activation of AMPK signalling in the hypothalamus increases feeding and reduces energy expenditure in the body [66].
Chronic administration of therapeutically relevant doses of clozapine, quetiapine, brexpiprazole, and lurasidone decreased IP3 synthesis, and increased AMP levels in the rat hypothalamus [46,47,67]. However, contrary to expectations, AMPK signallings were activated and unaffected by high-risk (clozapine and quetiapine) and low-risk (brexpiprazole and lurasidone) antipsychotics for weight gain, respectively [44,45,46,47,68]. Both clozapine and quetiapine are high-affinity antagonists of the histamine H1 receptor and the 5-HT2A receptor, whereas brexpiprazole and lurasidone are high-affinity 5-HT2A receptors but have low binding affinity to the H1 receptor [60,69]. Therefore, enhanced intra-hypothalamic AMPK signalling plays fundamental roles in antipsychotic-induced metabolic complications and weight gain, but decreasing IP3 with increasing AMP levels via inhibition of H1 and/or 5-HT2A receptors alone cannot explain the pathophysiology of antipsychotic-induced weight gain. Similar to clozapine, an H1 and 5-HT2A high-affinity atypical antipsychotic agent, olanzapine, which was established to also be a high-risk antipsychotic for weight gain, decreased IP3 synthesis [70,71]; however, olanzapine has been reported to enhance [61,72,73] and suppress [74,75] hypothalamic AMPK signalling with contradictory results. A recent study by Ferreira et al., which set the plasma concentration of olanzapine ranging from the maximum therapeutic concentration to the supratherapeutic level, demonstrated that olanzapine suppressed the hypothalamic AMPK signalling [75]. Considering that, clinically, olanzapine (lower than 10 mg/day) dose-dependently increased body weight, but above 10 mg/day, the weight gain induced by olanzapine displayed plateaus [59]; olanzapine has a dose-dependent biphasic effect on hypothalamic AMPK signalling, with activation by low-dose and suppression by high-dose. Further studies need to clarify these our hypothesis and the detailed mechanisms of dose-dependent biphasic action of olanzapine on AMPK signalling in the hypothalamus.

3. Clozapine and TRS

3.1. Efficacy of Clozapine in TRS

TRS is internationally defined by the Treatment Response and Resistance in Psychosis (TRRIP) Working Group and includes the following aspects: the presence of persistent symptoms—including positive and negative symptoms, and cognitive impairment—over at least 12 weeks of at least moderate severity caused by moderate levels of functional impairments [76]. Symptom classifications and thresholds should be based on standardised and validated clinical rating scales. Insufficient response to medication with at least two different antipsychotic medications, with a minimum treatment duration of twelve weeks (six weeks for each antipsychotic agent). This corresponds to a minimum dose equivalent to 600 mg per day of chlorpromazine. Confirmation of adequate treatment adherence is defined as the patient having taken at least 80% of the prescribed dose. To achieve this, at least two methods should be employed, including counting tablets, patient and caregiver reports, and review of medical records and documentation. Additionally, plasma drug concentrations should be monitored at least once for each antipsychotic agent [76,77].
Incontrovertible evidence supports the superior efficacy of clozapine compared with other atypical antipsychotics in improving positive symptoms and global psychopathology in TRS [5,13,78,79]. Considering the lack of evidence to support using polypharmacy of antipsychotics other than clozapine that is as effective as clozapine, the efficacy of clozapine in TRS is evaluated as being more robust [80]. Furthermore, patients treated with clozapine have also shown improvements in treatment adherence, resulting in decreased rehospitalisation rates [80,81].

3.2. Candidate Pathophysiology of TRS

Some research groups have emphasised the importance of distinguishing between primary and secondary TRS: primary TRS already presents with antipsychotic-resistant clinical features at the onset of the schizophrenia spectrum, whereas secondary TRS develops at later stages of the schizophrenia spectrum after an initial adequate response to antipsychotics [82,83,84]. Dopaminergic supersensitivity induced by consecutive exposure to antipsychotics has been speculated as a candidate mechanism of secondary TRS [85]. Persistent exposure to antipsychotics upregulates postsynaptic D2 receptors, leading to further psychotic exacerbation [85]. The estimated overall response rate to antipsychotic medications ranges from 40% to 60% [86,87]. The response rate to antipsychotic medication in antipsychotic-naïve patients is estimated to be approximately 75%; however, the response rate in a second trial using antipsychotic medications other than clozapine was considerably lower, ranging from 20% to 45% [88,89]. Response rates to clozapine have been reported to be maximally up to 80% when treatment is initiated within the first 2–3 years after resistance is established [87,88,90]. With subsequent initiation of clozapine medication, the response rate might be as low as 30% [87]. The efficacy of clozapine against TRS is significant compared with other antipsychotics but decreases depending on the duration of antipsychotic exposure, which is similar to other antipsychotics. These clinical findings regarding duration-dependent resistance at least partially support the dopaminergic supersensitivity hypothesis [85].
The specific features of clozapine, such as low affinity and rapid dissociation from D2 receptors, are considered to be candidate mechanisms via which clozapine-induced D2 receptor supersensitivity is less than that of other antipsychotics [20,38,69,91,92]. However, several line studies have demonstrated that the dissociation rate of clozapine from D2 receptors is not significantly faster compared with the rates of other antipsychotics, such as quetiapine, amisulpride, remoxipride, and sulpiride [20,93,94,95]. These pharmacodynamic findings suggest that the efficacy of clozapine in secondary TRS cannot be solely explained by either its low affinity or rapid dissociation from D2 receptors, even if the pathophysiology of TRS involves D2 receptor supersensitivity.

3.3. Candidate Targets of Clozapine Other Than Monoamine Receptors

Although schizophrenia is commonly speculated to be a pathophysiologically contiguous spectrum between treatment-responsive schizophrenia and TRS, several findings suggest that TRS might be a subtype with extreme characteristics from the perspective of neurodevelopmental disorders [96,97]. In other words, there are possibly two subtypes of pathophysiology of TRS, one being secondary treatment resistance due to long-term exposure to antipsychotic drugs, and the other already developing as TRS during the onset period. Approximately 70–80% of patients with TRS have been reported to present antipsychotic-resistant clinical features from the first episode [96,97]. Furthermore, predictors of antipsychotic resistance in schizophrenia are similar to the clinical features of ‘neurodevelopmental’ schizophrenia, such as being male, being of a younger age at onset, poor premorbid adjustment, and a longer duration of untreated illness [98,99]. So far, various studies have revealed impairments in cognitive components, such as sensorimotor function, attention, working memory, visuospatial processing, verbal intelligence, and memory in TRS patients compared with treatment-responsive schizophrenia [100,101,102,103]. These cognitive impairments are more suggestive of impaired function of glutamate transmission (via thalamocortical pathways) than monoamine transmission (via the mesolimbic and mesocortical systems). These cognitive impairment features of TRS suggest it may be caused by dysfunction of glutamatergic transmission (via thalamocortical pathways) rather than monoaminergic transmission (via mesolimbic and mesocortical pathways) [15,38,41,42,43,44,104].
Quantitative reviews of mRNA and protein expression of N-methyl-D-aspartate glutamate receptor (NMDA-R) in post-mortem studies have demonstrated that both mRNA and protein expression of the NR1 subunit of NMDA-R in the prefrontal cortex decreased in patients with schizophrenia compared with healthy volunteers [105]. mGluR5 (I-mGluR) signalling in the dorsolateral prefrontal cortex decreased, indicating that NMDA-R hypofunctions [106]. In the post-mortem frontal cortex of untreated patients with schizophrenia, downregulation of group II metabotropic glutamate receptors (II-mGluR), such as mGlu2/3, was reported [107]. Conversely, III-mGlu receptor expression in schizophrenia remains unreported, whereas the activation of the III-mGlu receptor suppressed the hyperactivated transmission induced by NMDA-R impairment in wild-type and II-mGluR deficit models [42,108].
In other line studies, both post-mortem and experimental animal model studies also demonstrated that impairment of the GABA-B receptor plays an important role in the pathophysiology of schizophrenia. Decreased GABA-B receptor expression in the hippocampus, prefrontal cortex, inferior temporal cortex, and entorhinal cortex in schizophrenia has been reported [109,110]. Decreased GABA-B receptor expression in the prefrontal cortex and hippocampus of the DBA/2J schizophrenia model compared with C57BL/6J mice was also revealed [111]. Clinically, clozapine is evaluated as the most effective antipsychotic to improve sensorimotor gating dysfunction in patients with schizophrenia [112]. Maladaptive perseveration with strategies that cannot lead to the desired outcome resulting from cognitive and behavioural inflexibility via possible sensorimotor gating dysfunction in the thalamocortical pathway is considered a characteristic feature of schizophrenia [38,60,69,113,114]. Pre-pulse inhibition (PPI) has been established as an endo-phenotype of sensorimotor gating function. Clozapine improved PPI deficits in an experimental animal model, ZFP804A mutant mice, and an NMDA/glutamate receptor (ketamine)-induced model [115,116]. Baclofen has also been indicated to counter PPI disruption of the acoustic startle reflex produced by the blockading of the NMDA-R [111]. Notably, the effects of baclofen on PPI deficit were comparable to those of clozapine but more prominent than those of the typical antipsychotic, haloperidol [111]. These behavioural studies suggest that the impacts of a GABA-B deficit contribute to sensorimotor impairment in schizophrenia.
A recent study using molecular docking calculations for the X-ray crystal structure of the GABA-B receptor suggested that clozapine, like baclofen, might bind to the GABA-B receptor [117]. Both clinical and preclinical studies have suggested that clozapine enhances GABA-B receptor function, and the direct binding of clozapine to the GABA-B receptor has not been demonstrated but, rather, has been denied [59,118,119]. Considering these previous findings, the enhancement of GABA-B receptor function with clozapine may be mediated by an indirect mechanism of clozapine rather than a direct agonist action. Therefore, the hypothesis regarding the stimulatory effects of clozapine on GABA-B receptor function is intriguing for understanding the underlying pathophysiology of the clinical efficacy of clozapine in TRS.

4. Impacts of L-BAIBA as the Pharmacodynamic Target of Clozapine

As mentioned above, the exploration of the pathophysiology of schizophrenia has developed over the last fifty years, whereas neither the mechanism of efficacy in TRS nor adverse reactions of clozapine have been clarified [120]. The characteristic mechanisms of clozapine absent in other atypical antipsychotics have not been elucidated [120].

4.1. Impacts of L-BAIBA on Metabolic Complications Induced by Clozapine

In 2012, we detected L-BAIBA release in the frontal cortex using microdialysis; however, a detailed release mechanism and function in the brain remain to be clarified [121]. Although the BAIBA enantiomer, a structural GABA isomer, was discovered in human urine in 1951 [122], its function has remained to be elucidated till the 2010s.
In the peripheral organs, BAIBA was rediscovered as a protective myokine that regulates adipose tissue browning, enhances insulin sensitivity, and improves obesity induced by a high-fat diet [123,124,125]. BAIBA increases Akt, AMPK, and insulin receptor substrate signalling and decreases the expression of gluconeogenic enzymes [125]. Activation of AMPK signalling is listed as a major therapeutic target for treating insulin-resistant diabetes [65,66]. Several clinical studies and meta-analyses have reported that antipsychotic-induced weight gain and metabolic complications are meaningfully improved and prevented with the AMPK activator metformin [53,55,126,127,128].
However, the activation of hypothalamic AMPK signalling may contribute to the pathophysiology of antipsychotic-induced weight gain and metabolic complications [46,60,61,64], since hypothalamic AMPK regulates both sides of the energy balance equation (feeding and energy expenditure) in the body [66]. Many pharmacodynamic studies have revealed that high-risk antipsychotics for weight gain and metabolic complications, such as clozapine, olanzapine, quetiapine, and zotepine activate AMPK signalling, but lower-risk antipsychotics, such as lurasidone and brexpiprazole, decrease and do not affect AMPK signalling in the hypothalamus and other brain regions, respectively [45,46,60,61,68,129,130]. Based on previous clinical and preclinical findings, it is hypothesised that clozapine activates hypothalamic signalling associated with AMPK via enhanced BAIBA signalling. The BAIBA enantiomer activates AMPK signalling in the hypothalamus and astrocytes [46,47,67] (Figure 2). According to our hypothesis, chronic administration of therapeutically relevant doses of clozapine increases the synthesis and release of the BAIBA enantiomer, but neither brexpiprazole nor lurasidone affects BAIBA in the hypothalamus [46,47,67]. This effect of clozapine on the BAIBA enantiomer is primarily on L-BAIBA, whereas the D-BAIBA level in the hypothalamus is lower than the limit of detection [46,47,67]. Therefore, L-BAIBA is a candidate molecule in the brain contributing to the pathophysiology of clozapine-induced weight gain and metabolic complications.

4.2. BAIBA Enantiomer Metabolism and Distribution

There are biologically two BAIBA enantiomers: D-BAIBA (R-BAIBA) and L-BAIBA (S-BAIBA) [131,132]. Although the structures of the BAIBA enantiomers are similar, their metabolic pathways function independently (Figure 3). D-BAIBA is synthesised from thymine and degraded by alanine-glyoxylate aminotransferase-2 [133]. Dihydropyrimidine dehydrogenase (DPYD) generates dihydrothymines from thymine [134]. Dihydropyrimidinase (DPYS) forms N-carbamoyl-β-aminoisobutyric acid (N-carbamoyl-BAIBA) from dihydrothymine. Finally, D-BAIBA is produced by beta-ureidopropionase (UPB1) from N-carbamoyl-BAIBA. This D-BAIBA synthesis process occurs in the cytosol, whereas D-BAIBA is depredated to D-methylmalonic semialdehyde (D-MMS) via glyoxylate aminotransferase 2 (AGXT2). Meanwhile, L-BAIBA is produced via the catabolism of the branched amino acid L-Valine in the mitochondria [135,136,137] (Figure 3). L-Valine is formed via ammonia and the oxidation reaction of methyl malonyl half aldehyde (L-methylmalonylsemialdehyde, L-MMS). L-MMS produces L-BAIBA in a reaction with the mitochondrial enzyme 4-aminobutyrate aminotransferase (ABAT) [138]. It has been reported that the production of L-BAIBA by ABAT is a bidirectional reaction, so the same enzyme can catalyse the conversion of L-BAIBA to L-MMS [135,139]. Furthermore, ABAT depredates GABA [140].
The literature on the distribution of D-BAIBA and L-BAIBA in plasma, urine, and tissues is contradictory. Most studies report that D-BAIBA is the main enantiomer of BAIBA in urine [132,141,142,143,144]. Another study suggested L-BAIBA is the major BAIBA enantiomer in the plasma [132], whereas others have reported that the more prevalent isoform is D-BAIBA [46,47,67,144,145]. Chronic administration of therapeutically relevant doses of clozapine increased L-BAIBA levels but did not affect D-BAIBA levels, resulting in unchanging overall plasma levels of the BAIBA enantiomer [46,47,67].

4.3. BAIBA Function in the CNS

Several functions of the BAIBA enantiomer in the CNS have been identified, such as the activation of glycine and GABA-A receptors [146,147]; however, the affinities of the BAIBA enantiomer to these receptors are relatively low, and subsequent functional analysis of the BAIBA enantiomer has not progressed. Clozapine also enhances the III-mGlu receptor and GABA-B receptors and denies direct binding to these receptors [42,108,117,148,149]. Considering that BAIBA is an isomer of GABA and is structurally similar, it is reasonable to predict that it may bind not only to GABA-A receptors but also to GABA-B receptors. The IC50 values of L-BAIBA to the III-mGlu receptor and GABA-B receptors are in approximately micromole orders [47].
The glycine receptor agonistic action of the BAIBA enantiomer can contribute to the interpretation of the pathophysiology of the effectiveness of clozapine in TRS. Various meta-analysis studies have elucidated that candidate NMDA-R modulators, including glycine, D-serine, N-acetyl-cysteine, and sarcosine, have exhibited favourable effects as augmentation therapy for atypical antipsychotics other than clozapine; however, when given to patients intaking clozapine, these modulators cannot improve but rather exacerbate schizophrenia symptoms [150,151,152].
Most neuroscientists have traditionally considered that primary information processing is implemented in the cortex and that the thalamus functions as a communication pathway for sensory input to the cortex [153]. Two signal transformation modes have been typically observed in thalamocortical glutamatergic neurones, bursting and tonic modes. The bursting mode is effective for detecting environmental changes, whereas the tonic mode is suitable for perceptual processing [154]. A well-known hypothesis regarding bottom-up cognition-promoting systems is that the hyperactivation of glutamatergic transmission in thalamocortical pathways plays important roles in several cognitive components, such as sensorimotor gaiting, sensory integration, and executive function [155]. Therefore, the thalamus probably plays an important role in implementing transformations between detection and perception modes [156]. The persistent tonic activation of thalamocortical projections has been observed in several models of schizophrenia, autism spectrum disorders, and attention-deficit/hyperactive disorder [42,60,69,130,157,158,159,160,161,162,163].
Systemic administration of NMDA-R antagonists drastically enhances thalamic glutamatergic neuronal activities via the inhibition of intra-thalamic GABAergic disinhibition (from the reticular thalamic nucleus to the mediodorsal thalamic nucleus, resulting in the activation of glutamatergic transmission from the thalamus to several cortex regions [42,47,158,164,165,166] (Figure 4)). Several studies have revealed that some atypical antipsychotics improve this tonic hyperactivation of thalamocortical glutamatergic transmission [38,41,43,44,45,46,47,60,67,68,69,114,129,130,159,166,167,168].
The local administration of several antipsychotics into the mediodorsal thalamic nucleus (MDTN) has been demonstrated to inhibit MK801-induced tonic activation of glutamatergic transmission in the thalamocortical pathway by suppressing glutamatergic neuronal activity in the MDTN [159,164,168]. In addition, local administration of clozapine into the mPFC suppresses MK801-induced hyper-glutamatergic transmission [42]. Studies have shown that the suppressive actions of clozapine on the tonic activation of thalamocortical glutamatergic transmission are predominant in the cortex rather than the thalamus, which is a distinguishing feature compared with other atypical antipsychotics [38,41,42,43].

5. Conclusions

Based on the accumulated recent clinical and preclinical findings, the present review introduced the possibility that L-BAIBA, a novel protective myokine in the peripheral organs, plays important roles in the mechanisms of the clinical actions of clozapine regarding its efficacy in treating TRS and its adverse reactions, such as weight gain and metabolic complications. In peripheral organs, L-BAIBA improves insulin-resistant diabetes by activating AMPK, and activated AMPK in the hypothalamus leads to weight gain. The suppression of IP3 synthesis through the inhibition of the H1 and 5-HT2A receptors has been considered a major mechanism of weight gain and metabolic complications associated with atypical antipsychotics. Low-risk atypical antipsychotics for weight gain (brexpiprazole and lurasidone) and clozapine decrease IP3 synthesis, leading to increasing intracellular AMP levels, whereas the effects on AMPK activity are different between low-risk antipsychotics for weight gain and clozapine. Both brexpiprazole and lurasidone do not activate AMPK signalling, but clozapine activates AMPK signalling in the hypothalamus. Therefore, L-BAIBA contributes to the pathophysiology of clozapine-induced metabolic complications via independent IP3 pathways. Persistent tonic hyperactivations of thalamocortical glutamatergic transmission produce sensorimotor deficits, which are considered a major component of cognitive impairment in TRS. Increasing L-BAIBA signalling in both the thalamus and prefrontal cortex attenuates the persistent tonic hyperactivations of thalamocortical glutamatergic transmission induced by NMDA-R disturbance via the activation of the GABA-A, GABA-B, glycine, and III-mGlu receptors. These recent preclinical findings suggest that the stimulatory effects of clozapine on L-BAIBA are, at least partially, involved in the mechanisms of clozapine’s clinical actions, such as its efficacy in TRS and metabolic complications.

Author Contributions

Conceptualization, M.O.; data curation, E.M., K.F. and M.O.; funding acquisition, M.O.; methodology, M.O.; project administration; M.O., validation, E.M. and M.O.; writing—original draft, M.O.; writing—review & editing, E.M., K.F. and M.O. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the Japan Society for the Promotion of Science (23K06986) and the Japan Epilepsy Research Foundation (JERF TENKAN 21008).

Institutional Review Board Statement

The study was approved by the Animal Research Ethics Committee of the Mie University School of Medicine (No. 2019-3, 31 October 2019).

Informed Consent Statement

Not applicable.

Data Availability Statement

The data that support the findings of this study are available from the corresponding author upon reasonable request. Some data may not be made available because of ethical restrictions.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Lally, J.; MacCabe, J.H. Antipsychotic medication in schizophrenia: A review. Br. Med. Bull. 2015, 114, 169–179. [Google Scholar] [CrossRef]
  2. Tandon, R. Schizophrenia and Other Psychotic Disorders in Diagnostic and Statistical Manual of Mental Disorders (DSM)-5: Clinical Implications of Revisions from DSM-IV. Indian J. Psychol. Med. 2014, 36, 223–225. [Google Scholar] [CrossRef] [PubMed]
  3. Tan, N.; Van Os, J. Schizofreniespectrum en andere psychotische stoornissen in de DSM-5. Tijdschr. Voor Psychiatr. 2014, 56, 167–172. [Google Scholar]
  4. Hu, R.J.; Malhotra, A.K.; Pickar, D. Predicting response to clozapine. CNS Drugs 1999, 11, 317–326. [Google Scholar] [CrossRef]
  5. Kane, J.; Honigfeld, G.; Singer, J.; Meltzer, H. Clozapine for the treatment-resistant schizophrenic: A double-blind comparison with chlorpromazine. Arch. Gen. Psychiatry 1988, 45, 789–796. [Google Scholar] [CrossRef]
  6. Meltzer, H.Y. Clozapine: Balancing safety with superior antipsychotic efficacy. Clin. Schizophr. Relat. Psychoses 2012, 6, 134–144. [Google Scholar] [CrossRef]
  7. Tiihonen, J.; Mittendorfer-Rutz, E.; Majak, M.; Mehtala, J.; Hoti, F.; Jedenius, E.; Enkusson, D.; Leval, A.; Sermon, J.; Tanskanen, A.; et al. Real-World Effectiveness of Antipsychotic Treatments in a Nationwide Cohort of 29823 Patients with Schizophrenia. JAMA Psychiatry 2017, 74, 686–693. [Google Scholar] [CrossRef] [PubMed]
  8. Falkai, P.; Wobrock, T.; Lieberman, J.; Glenthoj, B.; Gattaz, W.F.; Moller, H.J.; WFSBP Task Force on Treatment Guidelines for Schizophrenia. World Federation of Societies of Biological Psychiatry (WFSBP) guidelines for biological treatment of schizophrenia, part 2: Long-term treatment of schizophrenia. World J. Biol. Psychiatry 2006, 7, 5–40. [Google Scholar] [CrossRef]
  9. Hasan, A.; Falkai, P.; Wobrock, T.; Lieberman, J.; Glenthoj, B.; Gattaz, W.F.; Thibaut, F.; Moller, H.J.; WFSBP Task Force on Treatment Guidelines for Schizophrenia. World Federation of Societies of Biological Psychiatry (WFSBP) Guidelines for Biological Treatment of Schizophrenia, part 1: Update 2012 on the acute treatment of schizophrenia and the management of treatment resistance. World J. Biol. Psychiatry 2012, 13, 318–378. [Google Scholar] [CrossRef]
  10. Rogers, J.P.; Oldham, M.A.; Fricchione, G.; Northoff, G.; Ellen Wilson, J.; Mann, S.C.; Francis, A.; Wieck, A.; Elizabeth Wachtel, L.; Lewis, G.; et al. Evidence-based consensus guidelines for the management of catatonia: Recommendations from the British Association for Psychopharmacology. J. Psychopharmacol. 2023, 37, 327–369. [Google Scholar] [CrossRef]
  11. Masuda, T.; Misawa, F.; Takase, M.; Kane, J.M.; Correll, C.U. Association with Hospitalization and All-Cause Discontinuation Among Patients with Schizophrenia on Clozapine vs Other Oral Second-Generation Antipsychotics: A Systematic Review and Meta-analysis of Cohort Studies. JAMA Psychiatry 2019, 76, 1052. [Google Scholar] [CrossRef] [PubMed]
  12. Huhn, M.; Nikolakopoulou, A.; Schneider-Thoma, J.; Krause, M.; Samara, M.; Peter, N.; Arndt, T.; Backers, L.; Rothe, P.; Cipriani, A.; et al. Comparative efficacy and tolerability of 32 oral antipsychotics for the acute treatment of adults with multi-episode schizophrenia: A systematic review and network meta-analysis. Lancet 2019, 394, 939–951. [Google Scholar] [CrossRef] [PubMed]
  13. Mizuno, Y.; McCutcheon, R.A.; Brugger, S.P.; Howes, O.D. Heterogeneity and efficacy of antipsychotic treatment for schizophrenia with or without treatment resistance: A meta-analysis. Neuropsychopharmacology 2020, 45, 622–631. [Google Scholar] [CrossRef]
  14. Wenthur, C.J.; Lindsley, C.W. Classics in chemical neuroscience: Clozapine. ACS Chem. Neurosci. 2013, 4, 1018–1025. [Google Scholar] [CrossRef]
  15. Rusheen, A.E.; Gee, T.A.; Jang, D.P.; Blaha, C.D.; Bennet, K.E.; Lee, K.H.; Heien, M.L.; Oh, Y. Evaluation of electrochemical methods for tonic dopamine detection in vivo. TrAC Trends Anal. Chem. 2020, 132, 116049. [Google Scholar] [CrossRef]
  16. Meltzer, H.Y. Update on typical and atypical antipsychotic drugs. Annu. Rev. Med. 2013, 64, 393–406. [Google Scholar] [CrossRef]
  17. O’Connor, W.T.; O’Shea, S.D. Clozapine and GABA transmission in schizophrenia disease models: Establishing principles to guide treatments. Pharmacol. Ther. 2015, 150, 47–80. [Google Scholar] [CrossRef]
  18. Meltzer, H.Y.; Lee, M.A.; Ranjan, R.; Mason, E.A.; Cola, P.A. Relapse following clozapine withdrawal: Effect of neuroleptic drugs and cyproheptadine. Psychopharmacology 1996, 124, 176–187. [Google Scholar] [CrossRef]
  19. Correll, C.U. From receptor pharmacology to improved outcomes: Individualising the selection, dosing, and switching of antipsychotics. Eur. Psychiatry 2010, 25 (Suppl. S2), S12–S21. [Google Scholar] [CrossRef] [PubMed]
  20. Seeman, P. Clozapine, a fast-off-D2 antipsychotic. ACS Chem. Neurosci. 2014, 5, 24–29. [Google Scholar] [CrossRef] [PubMed]
  21. Su, T.P.; Malhotra, A.K.; Hadd, K.; Breier, A.; Pickar, D. D2 dopamine receptor occupancy: A crossover comparison of risperidone with clozapine therapy in schizophrenic patients. Arch. Gen. Psychiatry 1997, 54, 972–973. [Google Scholar] [CrossRef]
  22. Meltzer, H.Y. The mechanism of action of novel antipsychotic drugs. Schizophr. Bull. 1991, 17, 263–287. [Google Scholar] [CrossRef] [PubMed]
  23. Ishibashi, T.; Horisawa, T.; Tokuda, K.; Ishiyama, T.; Ogasa, M.; Tagashira, R.; Matsumoto, K.; Nishikawa, H.; Ueda, Y.; Toma, S.; et al. Pharmacological profile of lurasidone, a novel antipsychotic agent with potent 5-hydroxytryptamine 7 (5-HT7) and 5-HT1A receptor activity. J. Pharmacol. Exp. Ther. 2010, 334, 171–181. [Google Scholar] [CrossRef]
  24. Shapiro, D.A.; Renock, S.; Arrington, E.; Chiodo, L.A.; Liu, L.X.; Sibley, D.R.; Roth, B.L.; Mailman, R. Aripiprazole, a novel atypical antipsychotic drug with a unique and robust pharmacology. Neuropsychopharmacology 2003, 28, 1400–1411. [Google Scholar] [CrossRef]
  25. Ghanizadeh, A.; Sahraeizadeh, A.; Berk, M. A head-to-head comparison of aripiprazole and risperidone for safety and treating autistic disorders, a randomized double blind clinical trial. Child Psychiatry Hum. Dev. 2014, 45, 185–192. [Google Scholar] [CrossRef]
  26. Maeda, K.; Sugino, H.; Akazawa, H.; Amada, N.; Shimada, J.; Futamura, T.; Yamashita, H.; Ito, N.; McQuade, R.D.; Mork, A.; et al. Brexpiprazole I: In vitro and in vivo characterization of a novel serotonin-dopamine activity modulator. J. Pharmacol. Exp. Ther. 2014, 350, 589–604. [Google Scholar] [CrossRef] [PubMed]
  27. Fernandez, J.; Alonso, J.M.; Andres, J.I.; Cid, J.M.; Diaz, A.; Iturrino, L.; Gil, P.; Megens, A.; Sipido, V.K.; Trabanco, A.A. Discovery of new tetracyclic tetrahydrofuran derivatives as potential broad-spectrum psychotropic agents. J. Med. Chem. 2005, 48, 1709–1712. [Google Scholar] [CrossRef]
  28. Bymaster, F.; Calligaro, D.; Falcone, J.; Marsh, R.; Moore, N.; Tye, N.; Seeman, P.; Wong, D.J. Radioreceptor binding profile of the atypical antipsychotic olanzapine. Neuropsychopharmacology 1996, 14, 87–96. [Google Scholar] [CrossRef]
  29. Lopez-Munoz, F.; Alamo, C. Active metabolites as antidepressant drugs: The role of norquetiapine in the mechanism of action of quetiapine in the treatment of mood disorders. Front. Psychiatry 2013, 4, 102. [Google Scholar] [CrossRef] [PubMed]
  30. Smith, C.; Rahman, T.; Toohey, N.; Mazurkiewicz, J.; Herrick-Davis, K.; Teitler, M. Risperidone irreversibly binds to and inactivates the h5-HT7 serotonin receptor. Mol. Pharmacol. 2006, 70, 1264–1270. [Google Scholar] [CrossRef]
  31. Schotte, A.; Janssen, P.F.; Gommeren, W.; Luyten, W.H.; Van Gompel, P.; Lesage, A.S.; De Loore, K.; Leysen, J.E. Risperidone compared with new and reference antipsychotic drugs: In vitro and in vivo receptor binding. Psychopharmacology 1996, 124, 57–73. [Google Scholar] [CrossRef]
  32. Kroeze, W.K.; Hufeisen, S.J.; Popadak, B.A.; Renock, S.M.; Steinberg, S.; Ernsberger, P.; Jayathilake, K.; Meltzer, H.Y.; Roth, B.L. H1-histamine receptor affinity predicts short-term weight gain for typical and atypical antipsychotic drugs. Neuropsychopharmacology 2003, 28, 519–526. [Google Scholar] [CrossRef] [PubMed]
  33. Leysen, J.E.; Janssen, P.M.; Gommeren, W.; Wynants, J.; Pauwels, P.J.; Janssen, P.A. In vitro and in vivo receptor binding and effects on monoamine turnover in rat brain regions of the novel antipsychotics risperidone and ocaperidone. Mol. Pharmacol. 1992, 41, 494–508. [Google Scholar]
  34. Li, X.H.; Zhong, X.M.; Lu, L.; Zheng, W.; Wang, S.B.; Rao, W.W.; Wang, S.; Ng, C.H.; Ungvari, G.S.; Wang, G.; et al. The prevalence of agranulocytosis and related death in clozapine-treated patients: A comprehensive meta-analysis of observational studies. Psychol. Med. 2020, 50, 583–594. [Google Scholar] [CrossRef] [PubMed]
  35. Vickers, M.; Ramineni, V.; Malacova, E.; Eriksson, L.; McMahon, K.; Moudgil, V.; Scott, J.; Siskind, D. Risk factors for clozapine-induced myocarditis and cardiomyopathy: A systematic review and meta-analysis. Acta Psychiatr. Scand. 2022, 145, 442–455. [Google Scholar] [CrossRef]
  36. Pillinger, T.; McCutcheon, R.A.; Vano, L.; Mizuno, Y.; Arumuham, A.; Hindley, G.; Beck, K.; Natesan, S.; Efthimiou, O.; Cipriani, A.; et al. Comparative effects of 18 antipsychotics on metabolic function in patients with schizophrenia, predictors of metabolic dysregulation, and association with psychopathology: A systematic review and network meta-analysis. Lancet Psychiatry 2020, 7, 64–77. [Google Scholar] [CrossRef]
  37. De Berardis, D.; Rapini, G.; Olivieri, L.; Di Nicola, D.; Tomasetti, C.; Valchera, A.; Fornaro, M.; Di Fabio, F.; Perna, G.; Di Nicola, M.; et al. Safety of antipsychotics for the treatment of schizophrenia: A focus on the adverse effects of clozapine. Ther. Adv. Drug Saf. 2018, 9, 237–256. [Google Scholar] [CrossRef] [PubMed]
  38. Okada, M.; Fukuyama, K.; Shiroyama, T.; Murata, M. A Working Hypothesis Regarding Identical Pathomechanisms between Clinical Efficacy and Adverse Reaction of Clozapine via the Activation of Connexin43. Int. J. Mol. Sci. 2020, 21, 7019. [Google Scholar] [CrossRef] [PubMed]
  39. Blackman, G.; Oloyede, E. Clozapine discontinuation withdrawal symptoms in schizophrenia. Ther. Adv. Psychopharmacol. 2021, 11, 20451253211032053. [Google Scholar] [CrossRef]
  40. Lander, M.; Bastiampillai, T.; Sareen, J. Review of withdrawal catatonia: What does this reveal about clozapine? Transl. Psychiatry 2018, 8, 139. [Google Scholar] [CrossRef]
  41. Tanahashi, S.; Yamamura, S.; Nakagawa, M.; Motomura, E.; Okada, M. Clozapine, but not haloperidol, enhances glial D-serine and L-glutamate release in rat frontal cortex and primary cultured astrocytes. Br. J. Pharmacol. 2012, 165, 1543–1555. [Google Scholar] [CrossRef]
  42. Fukuyama, K.; Kato, R.; Murata, M.; Shiroyama, T.; Okada, M. Clozapine Normalizes a Glutamatergic Transmission Abnormality Induced by an Impaired NMDA Receptor in the Thalamocortical Pathway via the Activation of a Group III Metabotropic Glutamate Receptor. Biomolecules 2019, 9, 234. [Google Scholar] [CrossRef] [PubMed]
  43. Fukuyama, K.; Okubo, R.; Murata, M.; Shiroyama, T.; Okada, M. Activation of Astroglial Connexin is Involved in Concentration-Dependent Double-Edged Sword Clinical Action of Clozapine. Cells 2020, 9, 414. [Google Scholar] [CrossRef]
  44. Fukuyama, K.; Okada, M. Effects of Atypical Antipsychotics, Clozapine, Quetiapine and Brexpiprazole on Astroglial Transmission Associated with Connexin43. Int. J. Mol. Sci. 2021, 22, 5623. [Google Scholar] [CrossRef] [PubMed]
  45. Fukuyama, K.; Motomura, E.; Shiroyama, T.; Okada, M. Impact of 5-HT7 receptor inverse agonism of lurasidone on monoaminergic tripartite synaptic transmission and pathophysiology of lower risk of weight gain. Biomed. Pharmacother. 2022, 148, 112750. [Google Scholar] [CrossRef]
  46. Fukuyama, K.; Motomura, E.; Okada, M. Opposing effects of clozapine and brexpiprazole on beta-aminoisobutyric acid: Pathophysiology of antipsychotics-induced weight gain. Schizophrenia 2023, 9, 8. [Google Scholar] [CrossRef]
  47. Fukuyama, K.; Motomura, E.; Okada, M. Enhanced L-beta-Aminoisobutyric Acid Is Involved in the Pathophysiology of Effectiveness for Treatment-Resistant Schizophrenia and Adverse Reactions of Clozapine. Biomolecules 2023, 13, 862. [Google Scholar] [CrossRef]
  48. Kelly, A.C.; Sheitman, B.B.; Hamer, R.M.; Rhyne, D.C.; Reed, R.M.; Graham, K.A.; Rau, S.W.; Gilmore, J.H.; Perkins, D.O.; Peebles, S.S.; et al. A naturalistic comparison of the long-term metabolic adverse effects of clozapine versus other antipsychotics for patients with psychotic illnesses. J. Clin. Psychopharmacol. 2014, 34, 441–445. [Google Scholar] [CrossRef]
  49. Lund, B.C.; Perry, P.J.; Brooks, J.M.; Arndt, S. Clozapine use in patients with schizophrenia and the risk of diabetes, hyperlipidemia, and hypertension: A claims-based approach. Arch. Gen. Psychiatry 2001, 58, 1172–1176. [Google Scholar] [CrossRef] [PubMed]
  50. American Diabetes, A. Introduction: Standards of Medical Care in Diabetes-2022. Diabetes Care 2022, 45, S1–S2. [Google Scholar] [CrossRef]
  51. Taylor, S.I.; Yazdi, Z.S.; Beitelshees, A.L. Pharmacological treatment of hyperglycemia in type 2 diabetes. J. Clin. Investig. 2021, 131, e142243. [Google Scholar] [CrossRef] [PubMed]
  52. Popli, A.P.; Konicki, P.E.; Jurjus, G.J.; Fuller, M.A.; Jaskiw, G.E. Clozapine and associated diabetes mellitus. J. Clin. Psychiatry 1997, 58, 108–111. [Google Scholar] [CrossRef]
  53. Jiang, W.L.; Cai, D.B.; Yin, F.; Zhang, L.; Zhao, X.W.; He, J.; Ng, C.H.; Ungvari, G.S.; Sim, K.; Hu, M.L.; et al. Adjunctive metformin for antipsychotic-induced dyslipidemia: A meta-analysis of randomized, double-blind, placebo-controlled trials. Transl. Psychiatry 2020, 10, 117. [Google Scholar] [CrossRef]
  54. Zimbron, J.; Khandaker, G.M.; Toschi, C.; Jones, P.B.; Fernandez-Egea, E. A systematic review and meta-analysis of randomised controlled trials of treatments for clozapine-induced obesity and metabolic syndrome. Eur. Neuropsychopharmacol. 2016, 26, 1353–1365. [Google Scholar] [CrossRef] [PubMed]
  55. Siskind, D.J.; Leung, J.; Russell, A.W.; Wysoczanski, D.; Kisely, S. Metformin for Clozapine Associated Obesity: A Systematic Review and Meta-Analysis. PLoS ONE 2016, 11, e0156208. [Google Scholar] [CrossRef] [PubMed]
  56. Wang, C.; Shi, W.; Xu, J.; Huang, C.; Zhu, J. Outcomes and safety of concomitant topiramate or metformin for antipsychotics-induced obesity: A randomized-controlled trial. Ann. Gen. Psychiatry 2020, 19, 68. [Google Scholar] [CrossRef]
  57. Dayabandara, M.; Hanwella, R.; Ratnatunga, S.; Seneviratne, S.; Suraweera, C.; de Silva, V.A. Antipsychotic-associated weight gain: Management strategies and impact on treatment adherence. Neuropsychiatr. Dis. Treat. 2017, 13, 2231–2241. [Google Scholar] [CrossRef]
  58. Siskind, D.; Hahn, M.; Correll, C.U.; Fink-Jensen, A.; Russell, A.W.; Bak, N.; Broberg, B.V.; Larsen, J.; Ishoy, P.L.; Vilsboll, T.; et al. Glucagon-like peptide-1 receptor agonists for antipsychotic-associated cardio-metabolic risk factors: A systematic review and individual participant data meta-analysis. Diabetes Obes. Metab. 2019, 21, 293–302. [Google Scholar] [CrossRef]
  59. Wu, H.; Siafis, S.; Hamza, T.; Schneider-Thoma, J.; Davis, J.M.; Salanti, G.; Leucht, S. Antipsychotic-Induced Weight Gain: Dose-Response Meta-Analysis of Randomized Controlled Trials. Schizophr. Bull. 2022, 48, 643–654. [Google Scholar] [CrossRef]
  60. Fukuyama, K.; Motomura, E.; Okada, M. Therapeutic Potential and Limitation of Serotonin Type 7 Receptor Modulation. Int. J. Mol. Sci. 2023, 24, 2070. [Google Scholar] [CrossRef]
  61. Carli, M.; Kolachalam, S.; Longoni, B.; Pintaudi, A.; Baldini, M.; Aringhieri, S.; Fasciani, I.; Annibale, P.; Maggio, R.; Scarselli, M. Atypical Antipsychotics and Metabolic Syndrome: From Molecular Mechanisms to Clinical Differences. Pharmaceuticals (Basel) 2021, 14, 238. [Google Scholar] [CrossRef] [PubMed]
  62. Okada, M.; Yoshida, S.; Zhu, G.; Hirose, S.; Kaneko, S. Biphasic actions of topiramate on monoamine exocytosis associated with both soluble N-ethylmaleimide-sensitive factor attachment protein receptors and Ca(2+)-induced Ca(2+)-releasing systems. Neuroscience 2005, 134, 233–246. [Google Scholar] [CrossRef]
  63. de Brito, O.M.; Scorrano, L. An intimate liaison: Spatial organization of the endoplasmic reticulum-mitochondria relationship. EMBO J. 2010, 29, 2715–2723. [Google Scholar] [CrossRef]
  64. Decrock, E.; De Bock, M.; Wang, N.; Gadicherla, A.K.; Bol, M.; Delvaeye, T.; Vandenabeele, P.; Vinken, M.; Bultynck, G.; Krysko, D.V.; et al. IP3, a small molecule with a powerful message. Biochim. Biophys. Acta 2013, 1833, 1772–1786. [Google Scholar] [CrossRef] [PubMed]
  65. Foretz, M.; Guigas, B.; Bertrand, L.; Pollak, M.; Viollet, B. Metformin: From mechanisms of action to therapies. Cell Metab. 2014, 20, 953–966. [Google Scholar] [CrossRef]
  66. Lopez, M. Hypothalamic AMPK as a possible target for energy balance-related diseases. Trends Pharmacol. Sci. 2022, 43, 546–556. [Google Scholar] [CrossRef]
  67. Fukuyama, K.; Motomura, E.; Okada, M. A Candidate Gliotransmitter, L-β-Aminoisobutyrate, Contributes to Weight Gain and Metabolic Complication Induced by Atypical Antipsychotics. Nutrients 2023, 15, 1621. [Google Scholar] [CrossRef] [PubMed]
  68. Okada, M.; Fukuyama, K.; Motomura, E. Dose-Dependent Biphasic Action of Quetiapine on AMPK Signalling via 5-HT7 Receptor: Exploring Pathophysiology of Clinical and Adverse Effects of Quetiapine. Int. J. Mol. Sci. 2022, 23, 9103. [Google Scholar] [CrossRef]
  69. Okubo, R.; Hasegawa, T.; Fukuyama, K.; Shiroyama, T.; Okada, M. Current Limitations and Candidate Potential of 5-HT7 Receptor Antagonism in Psychiatric Pharmacotherapy. Front. Psychiatry 2021, 12, 623684. [Google Scholar] [CrossRef] [PubMed]
  70. Bymaster, F.P.; Nelson, D.L.; DeLapp, N.W.; Falcone, J.F.; Eckols, K.; Truex, L.L.; Foreman, M.M.; Lucaites, V.L.; Calligaro, D.O. Antagonism by olanzapine of dopamine D1, serotonin2, muscarinic, histamine H1 and alpha 1-adrenergic receptors in vitro. Schizophr. Res. 1999, 37, 107–122. [Google Scholar] [CrossRef]
  71. Arranz, B.; Rosel, P.; San, L.; Ramirez, N.; Duenas, R.M.; Salavert, J.; Centeno, M.; del Moral, E. Low baseline serotonin-2A receptors predict clinical response to olanzapine in first-episode schizophrenia patients. Psychiatry Res. 2007, 153, 103–109. [Google Scholar] [CrossRef]
  72. Deng, C.; Weston-Green, K.; Huang, X.F. The role of histaminergic H1 and H3 receptors in food intake: A mechanism for atypical antipsychotic-induced weight gain? Prog. Neuropsychopharmacol. Biol. Psychiatry 2010, 34, 1–4. [Google Scholar] [CrossRef] [PubMed]
  73. Han, M.; Lian, J.; Su, Y.; Deng, C. Cevimeline co-treatment attenuates olanzapine-induced metabolic disorders via modulating hepatic M3 muscarinic receptor: AMPKalpha signalling pathway in female rats. J. Psychopharmacol. 2022, 36, 202–213. [Google Scholar] [CrossRef]
  74. Castellani, L.N.; Pereira, S.; Kowalchuk, C.; Asgariroozbehani, R.; Singh, R.; Wu, S.; Hamel, L.; Alganem, K.; Ryan, W.G.; Zhang, X.; et al. Antipsychotics impair regulation of glucose metabolism by central glucose. Mol. Psychiatry 2022, 27, 4741–4753. [Google Scholar] [CrossRef]
  75. Ferreira, V.; Folgueira, C.; Guillen, M.; Zubiaur, P.; Navares, M.; Sarsenbayeva, A.; Lopez-Larrubia, P.; Eriksson, J.W.; Pereira, M.J.; Abad-Santos, F.; et al. Modulation of hypothalamic AMPK phosphorylation by olanzapine controls energy balance and body weight. Metabolism 2022, 137, 155335. [Google Scholar] [CrossRef] [PubMed]
  76. Howes, O.D.; McCutcheon, R.; Agid, O.; de Bartolomeis, A.; van Beveren, N.J.; Birnbaum, M.L.; Bloomfield, M.A.; Bressan, R.A.; Buchanan, R.W.; Carpenter, W.T.; et al. Treatment-Resistant Schizophrenia: Treatment Response and Resistance in Psychosis (TRRIP) Working Group Consensus Guidelines on Diagnosis and Terminology. Am. J. Psychiatry 2017, 174, 216–229. [Google Scholar] [CrossRef]
  77. Howes, O.D.; Thase, M.E.; Pillinger, T. Treatment resistance in psychiatry: State of the art and new directions. Mol. Psychiatry 2022, 27, 58–72. [Google Scholar] [CrossRef]
  78. Siskind, D.; McCartney, L.; Goldschlager, R.; Kisely, S. Clozapine v. first- and second-generation antipsychotics in treatment-refractory schizophrenia: Systematic review and meta-analysis. Br. J. Psychiatry 2016, 209, 385–392. [Google Scholar] [CrossRef] [PubMed]
  79. Wagner, E.; Siafis, S.; Fernando, P.; Falkai, P.; Honer, W.G.; Roh, A.; Siskind, D.; Leucht, S.; Hasan, A. Efficacy and safety of clozapine in psychotic disorders-a systematic quantitative meta-review. Transl. Psychiatry 2021, 11, 487. [Google Scholar] [CrossRef]
  80. Joo, S.W.; Kim, H.; Jo, Y.T.; Ahn, S.; Choi, Y.J.; Choi, W.; Park, S.; Lee, J. Comparative effectiveness of antipsychotic monotherapy and polypharmacy in schizophrenia patients with clozapine treatment: A nationwide, health insurance data-based study. Eur. Neuropsychopharmacol. 2022, 59, 36–44. [Google Scholar] [CrossRef]
  81. McEvoy, J.P.; Lieberman, J.A.; Stroup, T.S.; Davis, S.M.; Meltzer, H.Y.; Rosenheck, R.A.; Swartz, M.S.; Perkins, D.O.; Keefe, R.S.; Davis, C.E.; et al. Effectiveness of clozapine versus olanzapine, quetiapine, and risperidone in patients with chronic schizophrenia who did not respond to prior atypical antipsychotic treatment. Am. J. Psychiatry 2006, 163, 600–610. [Google Scholar] [CrossRef] [PubMed]
  82. Correll, C.U.; Howes, O.D. Treatment-Resistant Schizophrenia: Definition, Predictors, and Therapy Options. J. Clin. Psychiatry 2021, 82, MY20096AH1C. [Google Scholar] [CrossRef]
  83. Mouchlianitis, E.; McCutcheon, R.; Howes, O.D. Brain-imaging studies of treatment-resistant schizophrenia: A systematic review. Lancet Psychiatry 2016, 3, 451–463. [Google Scholar] [CrossRef]
  84. Potkin, S.G.; Kane, J.M.; Correll, C.U.; Lindenmayer, J.P.; Agid, O.; Marder, S.R.; Olfson, M.; Howes, O.D. The neurobiology of treatment-resistant schizophrenia: Paths to antipsychotic resistance and a roadmap for future research. NPJ Schizophr. 2020, 6, 1. [Google Scholar] [CrossRef] [PubMed]
  85. Wada, M.; Noda, Y.; Iwata, Y.; Tsugawa, S.; Yoshida, K.; Tani, H.; Hirano, Y.; Koike, S.; Sasabayashi, D.; Katayama, H.; et al. Dopaminergic dysfunction and excitatory/inhibitory imbalance in treatment-resistant schizophrenia and novel neuromodulatory treatment. Mol. Psychiatry 2022, 27, 2950–2967. [Google Scholar] [CrossRef] [PubMed]
  86. Siskind, D.; Siskind, V.; Kisely, S. Clozapine Response Rates among People with Treatment-Resistant Schizophrenia: Data from a Systematic Review and Meta-Analysis. Can. J. Psychiatry 2017, 62, 772–777. [Google Scholar] [CrossRef] [PubMed]
  87. Yoshimura, B.; Yada, Y.; So, R.; Takaki, M.; Yamada, N. The critical treatment window of clozapine in treatment-resistant schizophrenia: Secondary analysis of an observational study. Psychiatry Res. 2017, 250, 65–70. [Google Scholar] [CrossRef] [PubMed]
  88. Agid, O.; Arenovich, T.; Sajeev, G.; Zipursky, R.B.; Kapur, S.; Foussias, G.; Remington, G. An algorithm-based approach to first-episode schizophrenia: Response rates over 3 prospective antipsychotic trials with a retrospective data analysis. J. Clin. Psychiatry 2011, 72, 1439–1444. [Google Scholar] [CrossRef]
  89. Kahn, R.S.; Winter van Rossum, I.; Leucht, S.; McGuire, P.; Lewis, S.W.; Leboyer, M.; Arango, C.; Dazzan, P.; Drake, R.; Heres, S.; et al. Amisulpride and olanzapine followed by open-label treatment with clozapine in first-episode schizophrenia and schizophreniform disorder (OPTiMiSE): A three-phase switching study. Lancet Psychiatry 2018, 5, 797–807. [Google Scholar] [CrossRef] [PubMed]
  90. John, A.P.; Ko, E.K.F.; Dominic, A. Delayed Initiation of Clozapine Continues to Be a Substantial Clinical Concern. Can. J. Psychiatry 2018, 63, 526–531. [Google Scholar] [CrossRef]
  91. Kapur, S.; Seeman, P. Does fast dissociation from the dopamine d(2) receptor explain the action of atypical antipsychotics?: A new hypothesis. Am. J. Psychiatry 2001, 158, 360–369. [Google Scholar] [CrossRef] [PubMed]
  92. Stepnicki, P.; Kondej, M.; Kaczor, A.A. Current Concepts and Treatments of Schizophrenia. Molecules 2018, 23, 2087. [Google Scholar] [CrossRef] [PubMed]
  93. Sykes, D.A.; Moore, H.; Stott, L.; Holliday, N.; Javitch, J.A.; Lane, J.R.; Charlton, S.J. Extrapyramidal side effects of antipsychotics are linked to their association kinetics at dopamine D(2) receptors. Nat. Commun. 2017, 8, 763. [Google Scholar] [CrossRef] [PubMed]
  94. Sahlholm, K.; Zeberg, H.; Nilsson, J.; Ogren, S.O.; Fuxe, K.; Arhem, P. The fast-off hypothesis revisited: A functional kinetic study of antipsychotic antagonism of the dopamine D2 receptor. Eur. Neuropsychopharmacol. 2016, 26, 467–476. [Google Scholar] [CrossRef]
  95. Kapur, S.; Seeman, P. Antipsychotic agents differ in how fast they come off the dopamine D2 receptors. Implications for atypical antipsychotic action. J. Psychiatry Neurosci. 2000, 25, 161–166. [Google Scholar]
  96. Demjaha, A.; Lappin, J.M.; Stahl, D.; Patel, M.X.; MacCabe, J.H.; Howes, O.D.; Heslin, M.; Reininghaus, U.A.; Donoghue, K.; Lomas, B.; et al. Antipsychotic treatment resistance in first-episode psychosis: Prevalence, subtypes and predictors. Psychol. Med. 2017, 47, 1981–1989. [Google Scholar] [CrossRef] [PubMed]
  97. Lally, J.; Ajnakina, O.; Di Forti, M.; Trotta, A.; Demjaha, A.; Kolliakou, A.; Mondelli, V.; Reis Marques, T.; Pariante, C.; Dazzan, P.; et al. Two distinct patterns of treatment resistance: Clinical predictors of treatment resistance in first-episode schizophrenia spectrum psychoses. Psychol. Med. 2016, 46, 3231–3240. [Google Scholar] [CrossRef] [PubMed]
  98. Carbon, M.; Correll, C.U. Clinical predictors of therapeutic response to antipsychotics in schizophrenia. Dialogues Clin. Neurosci. 2014, 16, 505–524. [Google Scholar] [CrossRef]
  99. Murray, R.M.; O’Callaghan, E.; Castle, D.J.; Lewis, S.W. A neurodevelopmental approach to the classification of schizophrenia. Schizophr. Bull. 1992, 18, 319–332. [Google Scholar] [CrossRef]
  100. Bourque, J.; Lakis, N.; Champagne, J.; Stip, E.; Lalonde, P.; Lipp, O.; Mendrek, A. Clozapine and visuospatial processing in treatment-resistant schizophrenia. Cogn. Neuropsychiatry 2013, 18, 615–630. [Google Scholar] [CrossRef]
  101. de Bartolomeis, A.; Balletta, R.; Giordano, S.; Buonaguro, E.F.; Latte, G.; Iasevoli, F. Differential cognitive performances between schizophrenic responders and non-responders to antipsychotics: Correlation with course of the illness, psychopathology, attitude to the treatment and antipsychotics doses. Psychiatry Res. 2013, 210, 387–395. [Google Scholar] [CrossRef] [PubMed]
  102. Huang, J.; Zhu, Y.; Fan, F.; Chen, S.; Hong, Y.; Cui, Y.; Luo, X.; Tan, S.; Wang, Z.; Shang, L.; et al. Hippocampus and cognitive domain deficits in treatment-resistant schizophrenia: A comparison with matched treatment-responsive patients and healthy controls(✰,✰✰, bigstar, bigstar bigstar). Psychiatry Res. Neuroimaging 2020, 297, 111043. [Google Scholar] [CrossRef] [PubMed]
  103. Lin, A.S.; Chan, H.Y.; Peng, Y.C.; Chen, W.J. Severity in sustained attention impairment and clozapine-resistant schizophrenia: A retrospective study. BMC Psychiatry 2019, 19, 220. [Google Scholar] [CrossRef]
  104. Ito, Y.; Murata, M.; Taku, O.; Fukuyama, K.; Motomura, E.; Dohi, K.; Okada, M. Developed catatonia with rhabdomyolysis and exacerbated cardiac failure upon switching from clozapine to olanzapine owing to cardiomyopathy during clozapine medication—A case report. Asian J. Psychiatr. 2023, 80, 103376. [Google Scholar] [CrossRef] [PubMed]
  105. Kruse, A.O.; Bustillo, J.R. Glutamatergic dysfunction in Schizophrenia. Transl. Psychiatry 2022, 12, 500. [Google Scholar] [CrossRef]
  106. Wang, H.Y.; MacDonald, M.L.; Borgmann-Winter, K.E.; Banerjee, A.; Sleiman, P.; Tom, A.; Khan, A.; Lee, K.C.; Roussos, P.; Siegel, S.J.; et al. mGluR5 hypofunction is integral to glutamatergic dysregulation in schizophrenia. Mol. Psychiatry 2020, 25, 750–760. [Google Scholar] [CrossRef]
  107. Gonzalez-Maeso, J.; Ang, R.L.; Yuen, T.; Chan, P.; Weisstaub, N.V.; Lopez-Gimenez, J.F.; Zhou, M.; Okawa, Y.; Callado, L.F.; Milligan, G.; et al. Identification of a serotonin/glutamate receptor complex implicated in psychosis. Nature 2008, 452, 93–97. [Google Scholar] [CrossRef]
  108. Fell, M.J.; Svensson, K.A.; Johnson, B.G.; Schoepp, D.D. Evidence for the role of metabotropic glutamate (mGlu)2 not mGlu3 receptors in the preclinical antipsychotic pharmacology of the mGlu2/3 receptor agonist (-)-(1R,4S,5S,6S)-4-amino-2-sulfonylbicyclo[3.1.0]hexane-4,6-dicarboxylic acid (LY404039). J. Pharmacol. Exp. Ther. 2008, 326, 209–217. [Google Scholar] [CrossRef]
  109. Mizukami, K.; Sasaki, M.; Ishikawa, M.; Iwakiri, M.; Hidaka, S.; Shiraishi, H.; Iritani, S. Immunohistochemical localization of gamma-aminobutyric acid(B) receptor in the hippocampus of subjects with schizophrenia. Neurosci. Lett. 2000, 283, 101–104. [Google Scholar] [CrossRef]
  110. Ishikawa, M.; Mizukami, K.; Iwakiri, M.; Asada, T. Immunohistochemical and immunoblot analysis of gamma-aminobutyric acid B receptor in the prefrontal cortex of subjects with schizophrenia and bipolar disorder. Neurosci. Lett. 2005, 383, 272–277. [Google Scholar] [CrossRef]
  111. Bortolato, M.; Frau, R.; Orru, M.; Piras, A.P.; Fa, M.; Tuveri, A.; Puligheddu, M.; Gessa, G.L.; Castelli, M.P.; Mereu, G.; et al. Activation of GABA(B) receptors reverses spontaneous gating deficits in juvenile DBA/2J mice. Psychopharmacology 2007, 194, 361–369. [Google Scholar] [CrossRef] [PubMed]
  112. Micoulaud-Franchi, J.A.; Aramaki, M.; Geoffroy, P.A.; Richieri, R.; Cermolacce, M.; Faget, C.; Ystad, S.; Kronland-Martinet, R.; Lancon, C.; Vion-Dury, J. Effects of clozapine on perceptual abnormalities and sensory gating: A preliminary cross-sectional study in schizophrenia. J. Clin. Psychopharmacol. 2015, 35, 184–187. [Google Scholar] [CrossRef] [PubMed]
  113. Beas, B.S.; Setlow, B.; Bizon, J.L. Effects of acute administration of the GABA(B) receptor agonist baclofen on behavioral flexibility in rats. Psychopharmacology 2016, 233, 2787–2797. [Google Scholar] [CrossRef]
  114. Okada, M.; Kawano, Y.; Fukuyama, K.; Motomura, E.; Shiroyama, T. Candidate Strategies for Development of a Rapid-Acting Antidepressant Class That Does Not Result in Neuropsychiatric Adverse Effects: Prevention of Ketamine-Induced Neuropsychiatric Adverse Reactions. Int. J. Mol. Sci. 2020, 21, 7951. [Google Scholar] [CrossRef] [PubMed]
  115. Huang, Y.; Huang, J.; Zhou, Q.X.; Yang, C.X.; Yang, C.P.; Mei, W.Y.; Zhang, L.; Zhang, Q.; Hu, L.; Hu, Y.Q.; et al. ZFP804A mutant mice display sex-dependent schizophrenia-like behaviors. Mol. Psychiatry 2021, 26, 2514–2532. [Google Scholar] [CrossRef] [PubMed]
  116. Yang, S.Y.; Hong, C.J.; Huang, Y.H.; Tsai, S.J. The effects of glycine transporter I inhibitor, N-methylglycine (sarcosine), on ketamine-induced alterations in sensorimotor gating and regional brain c-Fos expression in rats. Neurosci. Lett. 2010, 469, 127–130. [Google Scholar] [CrossRef] [PubMed]
  117. Nair, P.C.; McKinnon, R.A.; Miners, J.O.; Bastiampillai, T. Binding of clozapine to the GABAB receptor: Clinical and structural insights. Mol. Psychiatry 2020, 25, 1910–1919. [Google Scholar] [CrossRef]
  118. Roth BL, D.J. Psychoactive Drug Screening Program (PDSP). 2017. Available online: https://pdsp.unc.edu/databases/pdsp.php?knowID=0&kiKey=&receptorDD=&receptor=&speciesDD=&species=&sourcesDD=&source=&hotLigandDD=&hotLigand=&testLigandDD=&testFreeRadio=testFreeRadio&testLigand=clozapine&referenceDD=&reference=&KiGreater=&KiLess=&kiAllRadio=all&doQuery=Submit+Query (accessed on 27 July 2023).
  119. Miyazawa, A.; Kanahara, N.; Shiko, Y.; Ozawa, Y.; Kawasaki, Y.; Komatsu, H.; Masumo, Y.; Nakata, Y.; Iyo, M. The cortical silent period in schizophrenia: A systematic review and meta-analysis focusing on disease stage and antipsychotic medication. J. Psychopharmacol. 2022, 36, 479–488. [Google Scholar] [CrossRef]
  120. de Bartolomeis, A.; Vellucci, L.; Barone, A.; Manchia, M.; De Luca, V.; Iasevoli, F.; Correll, C.U. Clozapine’s multiple cellular mechanisms: What do we know after more than fifty years? A systematic review and critical assessment of translational mechanisms relevant for innovative strategies in treatment-resistant schizophrenia. Pharmacol. Ther. 2022, 236, 108236. [Google Scholar] [CrossRef]
  121. Tanahashi, S.; Ueda, Y.; Nakajima, A.; Yamamura, S.; Nagase, H.; Okada, M. Novel delta1-receptor agonist KNT-127 increases the release of dopamine and L-glutamate in the striatum, nucleus accumbens and median pre-frontal cortex. Neuropharmacology 2012, 62, 2057–2067. [Google Scholar] [CrossRef]
  122. Crumpler, H.R.; Dent, C.E.; Harris, H.; Westall, R.G. beta-Aminoisobutyric acid (alpha-methyl-beta-alanine); a new amino-acid obtained from human urine. Nature 1951, 167, 307–308. [Google Scholar] [CrossRef]
  123. Roberts, L.D.; Bostrom, P.; O’Sullivan, J.F.; Schinzel, R.T.; Lewis, G.D.; Dejam, A.; Lee, Y.K.; Palma, M.J.; Calhoun, S.; Georgiadi, A.; et al. beta-Aminoisobutyric acid induces browning of white fat and hepatic beta-oxidation and is inversely correlated with cardiometabolic risk factors. Cell Metab. 2014, 19, 96–108. [Google Scholar] [CrossRef]
  124. Jung, T.W.; Hwang, H.J.; Hong, H.C.; Yoo, H.J.; Baik, S.H.; Choi, K.M. BAIBA attenuates insulin resistance and inflammation induced by palmitate or a high fat diet via an AMPK-PPARdelta-dependent pathway in mice. Diabetologia 2015, 58, 2096–2105. [Google Scholar] [CrossRef] [PubMed]
  125. Shi, C.X.; Zhao, M.X.; Shu, X.D.; Xiong, X.Q.; Wang, J.J.; Gao, X.Y.; Chen, Q.; Li, Y.H.; Kang, Y.M.; Zhu, G.Q. beta-aminoisobutyric acid attenuates hepatic endoplasmic reticulum stress and glucose/lipid metabolic disturbance in mice with type 2 diabetes. Sci. Rep. 2016, 6, 21924. [Google Scholar] [CrossRef]
  126. Zheng, W.; Li, X.B.; Tang, Y.L.; Xiang, Y.Q.; Wang, C.Y.; de Leon, J. Metformin for Weight Gain and Metabolic Abnormalities Associated with Antipsychotic Treatment: Meta-Analysis of Randomized Placebo-Controlled Trials. J. Clin. Psychopharmacol. 2015, 35, 499–509. [Google Scholar] [CrossRef] [PubMed]
  127. de Silva, V.A.; Suraweera, C.; Ratnatunga, S.S.; Dayabandara, M.; Wanniarachchi, N.; Hanwella, R. Metformin in prevention and treatment of antipsychotic induced weight gain: A systematic review and meta-analysis. BMC Psychiatry 2016, 16, 341. [Google Scholar] [CrossRef]
  128. Zheng, W.; Zhang, Q.E.; Cai, D.B.; Yang, X.H.; Ungvari, G.S.; Ng, C.H.; Wu, R.R.; Xiang, Y.T. Combination of Metformin and Lifestyle Intervention for Antipsychotic-Related Weight Gain: A Meta-Analysis of Randomized Controlled Trials. Pharmacopsychiatry 2019, 52, 24–31. [Google Scholar] [CrossRef] [PubMed]
  129. Fukuyama, K.; Motomura, E.; Okada, M. Brexpiprazole reduces 5-HT7 receptor function on astroglial transmission systems. Int. J. Mol. Sci. 2022, 23, 6571. [Google Scholar] [CrossRef]
  130. Fukuyama, K.; Okada, M. Effects of an Atypical Antipsychotic, Zotepine, on Astroglial L-Glutamate Release through Hemichannels: Exploring the Mechanism of Mood-Stabilising Antipsychotic Actions and Antipsychotic-Induced Convulsion. Pharmaceuticals 2021, 14, 1116. [Google Scholar] [CrossRef]
  131. Vemula, H.; Kitase, Y.; Ayon, N.J.; Bonewald, L.; Gutheil, W.G. Gaussian and linear deconvolution of LC-MS/MS chromatograms of the eight aminobutyric acid isomers. Anal. Biochem. 2017, 516, 75–85. [Google Scholar] [CrossRef]
  132. Solem, E.; Jellum, E.; Eldjarn, L. The absolute configuration of β-aminoisobutyric acid in human serum and urine. Clinica Chimica Acta 1974, 50, 393–403. [Google Scholar] [CrossRef]
  133. Lee, I.S.; Nishikimi, M.; Inoue, M.; Muragaki, Y.; Ooshima, A. Specific expression of alanine-glyoxylate aminotransferase 2 in the epithelial cells of Henle’s loop. Nephron 1999, 83, 184–185. [Google Scholar] [CrossRef] [PubMed]
  134. Fink, K.; Cline, R.E.; Henderson, R.B.; Fink, R.M. Metabolism of thymine (methyl-C14 or -2-C14) by rat liver in vitro. J. Biol. Chem. 1956, 221, 425–433. [Google Scholar] [CrossRef] [PubMed]
  135. Roe, C.R.; Struys, E.; Kok, R.M.; Roe, D.S.; Harris, R.A.; Jakobs, C. Methylmalonic semialdehyde dehydrogenase deficiency: Psychomotor delay and methylmalonic aciduria without metabolic decompensation. Mol. Genet. Metab. 1998, 65, 35–43. [Google Scholar] [CrossRef] [PubMed]
  136. Pollitt, R.J.; Green, A.; Smith, R. Excessive excretion of beta-alanine and of 3-hydroxypropionic, R- and S-3-aminoisobutyric, R- and S-3-hydroxyisobutyric and S-2-(hydroxymethyl)butyric acids probably due to a defect in the metabolism of the corresponding malonic semialdehydes. J. Inherit. Metab. Dis. 1985, 8, 75–79. [Google Scholar] [CrossRef]
  137. Kakimoto, Y.; Kanazawa, A.; Taniguchi, K.; Sano, I. Beta-aminoisobutyrate-alpha-ketoglutarate transaminase in relation to beta-aminoisobutyric aciduria. Biochim. Biophys. Acta 1968, 156, 374–380. [Google Scholar] [CrossRef]
  138. Kamei, Y.; Hatazawa, Y.; Uchitomi, R.; Yoshimura, R.; Miura, S. Regulation of Skeletal Muscle Function by Amino Acids. Nutrients 2020, 12, 261. [Google Scholar] [CrossRef]
  139. Tamaki, N.; Kaneko, M.; Kikugawa, M.; Fujimoto, S. Evaluation of interconversion between (R)- and (S)-enantiomers of beta-aminoisobutyrate. Biochim. Biophys. Acta 1990, 1035, 117–119. [Google Scholar] [CrossRef]
  140. Koenig, M.K.; Hodgeman, R.; Riviello, J.J.; Chung, W.; Bain, J.; Chiriboga, C.A.; Ichikawa, K.; Osaka, H.; Tsuji, M.; Gibson, K.M.J.N. Phenotype of GABA-transaminase deficiency. Neurology 2017, 88, 1919–1924. [Google Scholar] [CrossRef]
  141. van Gennip, A.H.; Kamerling, J.P.; de Bree, P.K.; Wadman, S.K. Linear relationship between the R- and S-enantiomers of a beta-aminoisobutyric acid in human urine. Clin. Chim. Acta 1981, 116, 261–267. [Google Scholar] [CrossRef]
  142. Armstrong, M.D.; Yates, K.; Kakimoto, Y.; Taniguchi, K.; Kappe, T. Excretion of β-aminoisobutyric acid by man. J. Biol. Chem. 1963, 238, 1447–1455. [Google Scholar] [CrossRef]
  143. Gejyo, F.; Kinoshita, Y.; Ikenaka, T. Identification of beta-aminoisobutyric acid in uremic serum. Clin. Chim. Acta 1976, 70, 407–415. [Google Scholar] [CrossRef] [PubMed]
  144. Stautemas, J.; Van Kuilenburg, A.B.P.; Stroomer, L.; Vaz, F.; Blancquaert, L.; Lefevere, F.B.D.; Everaert, I.; Derave, W. Acute Aerobic Exercise Leads to Increased Plasma Levels of R- and S-beta-Aminoisobutyric Acid in Humans. Front. Physiol. 2019, 10, 1240. [Google Scholar] [CrossRef] [PubMed]
  145. Tanianskii, D.A.; Jarzebska, N.; Birkenfeld, A.L.; O’Sullivan, J.F.; Rodionov, R.N. Beta-Aminoisobutyric Acid as a Novel Regulator of Carbohydrate and Lipid Metabolism. Nutrients 2019, 11, 524. [Google Scholar] [CrossRef] [PubMed]
  146. Horikoshi, T.; Asanuma, A.; Yanagisawa, K.; Anzai, K.; Goto, S. Taurine and beta-alanine act on both GABA and glycine receptors in Xenopus oocyte injected with mouse brain messenger RNA. Brain Res. 1988, 464, 97–105. [Google Scholar] [CrossRef]
  147. Schmieden, V.; Betz, H. Pharmacology of the inhibitory glycine receptor: Agonist and antagonist actions of amino acids and piperidine carboxylic acid compounds. Mol. Pharmacol. 1995, 48, 919–927. [Google Scholar]
  148. de la Fuente Revenga, M.; Ibi, D.; Cuddy, T.; Toneatti, R.; Kurita, M.; Ijaz, M.K.; Miles, M.F.; Wolstenholme, J.T.; Gonzalez-Maeso, J. Chronic clozapine treatment restrains via HDAC2 the performance of mGlu2 receptor agonism in a rodent model of antipsychotic activity. Neuropsychopharmacology 2019, 44, 443–454. [Google Scholar] [CrossRef]
  149. Wu, Y.; Blichowski, M.; Daskalakis, Z.J.; Wu, Z.; Liu, C.C.; Cortez, M.A.; Snead, O.C., 3rd. Evidence that clozapine directly interacts on the GABAB receptor. Neuroreport 2011, 22, 637–641. [Google Scholar] [CrossRef]
  150. Goh, K.K.; Wu, T.H.; Chen, C.H.; Lu, M.L. Efficacy of N-methyl-D-aspartate receptor modulator augmentation in schizophrenia: A meta-analysis of randomised, placebo-controlled trials. J. Psychopharmacol. 2021, 35, 236–252. [Google Scholar] [CrossRef]
  151. Marchi, M.; Galli, G.; Magarini, F.M.; Mattei, G.; Galeazzi, G.M. Sarcosine as an add-on treatment to antipsychotic medication for people with schizophrenia: A systematic review and meta-analysis of randomized controlled trials. Expert. Opin. Drug Metab. Toxicol. 2021, 17, 483–493. [Google Scholar] [CrossRef]
  152. Singh, S.P.; Singh, V. Meta-analysis of the efficacy of adjunctive NMDA receptor modulators in chronic schizophrenia. CNS Drugs 2011, 25, 859–885. [Google Scholar] [CrossRef] [PubMed]
  153. Basso, M.A.; Uhlrich, D.; Bickford, M.E. Cortical function: A view from the thalamus. Neuron 2005, 45, 485–488. [Google Scholar] [CrossRef] [PubMed]
  154. Groh, A.; Bokor, H.; Mease, R.A.; Plattner, V.M.; Hangya, B.; Stroh, A.; Deschenes, M.; Acsady, L. Convergence of cortical and sensory driver inputs on single thalamocortical cells. Cereb. Cortex 2014, 24, 3167–3179. [Google Scholar] [CrossRef] [PubMed]
  155. Cruz, K.G.; Leow, Y.N.; Le, N.M.; Adam, E.; Huda, R.; Sur, M. Cortical-subcortical interactions in goal-directed behavior. Physiol. Rev. 2023, 103, 347–389. [Google Scholar] [CrossRef]
  156. Ahissar, E.; Oram, T. Thalamic relay or cortico-thalamic processing? Old question, new answers. Cereb. Cortex 2015, 25, 845–848. [Google Scholar] [CrossRef]
  157. Okada, M.; Fukuyama, K.; Kawano, Y.; Shiroyama, T.; Suzuki, D.; Ueda, Y. Effects of acute and sub-chronic administrations of guanfacine on catecholaminergic transmissions in the orbitofrontal cortex. Neuropharmacology 2019, 156, 107547. [Google Scholar] [CrossRef]
  158. Okada, M.; Fukuyama, K.; Okubo, R.; Shiroyama, T.; Ueda, Y. Lurasidone Sub-Chronically Activates Serotonergic Transmission via Desensitization of 5-HT1A and 5-HT7 Receptors in Dorsal Raphe Nucleus. Pharmaceuticals 2019, 12, 149. [Google Scholar] [CrossRef]
  159. Okada, M.; Fukuyama, K.; Ueda, Y. Lurasidone inhibits NMDA receptor antagonist-induced functional abnormality of thalamocortical glutamatergic transmission via 5-HT7 receptor blockade. Br. J. Pharmacol. 2019, 176, 4002–4018. [Google Scholar] [CrossRef]
  160. Fukuyama, K.; Fukuzawa, M.; Shiroyama, T.; Okada, M. Pathogenesis and pathophysiology of autosomal dominant sleep-related hypermotor epilepsy with S284L-mutant alpha4 subunit of nicotinic ACh receptor. Br. J. Pharmacol. 2020, 177, 2143–2162. [Google Scholar] [CrossRef]
  161. Fukuyama, K.; Okada, M. Age-Dependent and Sleep/Seizure-Induced Pathomechanisms of Autosomal Dominant Sleep-Related Hypermotor Epilepsy. Int. J. Mol. Sci. 2020, 21, 8142. [Google Scholar] [CrossRef]
  162. Okada, M. Can rodent models elucidate the pathomechanisms of genetic epilepsy? Br. J. Pharmacol. 2022, 179, 1620–1639. [Google Scholar] [CrossRef] [PubMed]
  163. Fukuyama, K.; Nakano, T.; Shiroyama, T.; Okada, M. Chronic Administrations of Guanfacine on Mesocortical Catecholaminergic and Thalamocortical Glutamatergic Transmissions. Int. J. Mol. Sci. 2021, 22, 4122. [Google Scholar] [CrossRef]
  164. Fukuyama, K.; Hasegawa, T.; Okada, M. Cystine/Glutamate Antiporter and Aripiprazole Compensate NMDA Antagonist-Induced Dysfunction of Thalamocortical L-Glutamatergic Transmission. Int. J. Mol. Sci. 2018, 19, 3645. [Google Scholar] [CrossRef] [PubMed]
  165. Okada, M.; Okubo, R.; Fukuyama, K. Vortioxetine Subchronically Activates Serotonergic Transmission via Desensitization of Serotonin 5-HT1A Receptor with 5-HT3 Receptor Inhibition in Rats. Int. J. Mol. Sci. 2019, 20, 6235. [Google Scholar] [CrossRef] [PubMed]
  166. Okada, M.; Fukuyama, K. Interaction between Mesocortical and Mesothalamic Catecholaminergic Transmissions Associated with NMDA Receptor in the Locus Coeruleus. Biomolecules 2020, 10, 990. [Google Scholar] [CrossRef] [PubMed]
  167. Okada, M.; Oka, T.; Nakamoto, M.; Fukuyama, K.; Shiroyama, T. Astroglial Connexin43 as a Potential Target for a Mood Stabiliser. Int. J. Mol. Sci. 2020, 22, 339. [Google Scholar] [CrossRef] [PubMed]
  168. Okada, M.; Matsumoto, R.; Yamamoto, Y.; Fukuyama, K. Effects of Subchronic Administrations of Vortioxetine, Lurasidone, and Escitalopram on Thalamocortical Glutamatergic Transmission Associated with Serotonin 5-HT7 Receptor. Int. J. Mol. Sci. 2021, 22, 1351. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic presentation of hypothalamic signalling associated with traditional hypothesis regarding the mechanisms of antipsychotic-induced metabolic complications and weight gain. Red and blue arrows indicate activation and inhibition, respectively. Abbreviations: H1 receptor—histamine H1 receptor, 5-HT2A receptor—serotonin 5-HT2A receptor, IP3—of inositol trisphosphate, CICR—Ca2+-induced Ca2+-releasing system, ATP—adenosine triphosphate, AMP—adenosine monophosphate, and AMPK—AMP-activated protein kinase.
Figure 1. Schematic presentation of hypothalamic signalling associated with traditional hypothesis regarding the mechanisms of antipsychotic-induced metabolic complications and weight gain. Red and blue arrows indicate activation and inhibition, respectively. Abbreviations: H1 receptor—histamine H1 receptor, 5-HT2A receptor—serotonin 5-HT2A receptor, IP3—of inositol trisphosphate, CICR—Ca2+-induced Ca2+-releasing system, ATP—adenosine triphosphate, AMP—adenosine monophosphate, and AMPK—AMP-activated protein kinase.
Biomolecules 13 01288 g001
Figure 2. Proposed hypothesis about the mechanism of clozapine-induced metabolic complication of weight gain associated with enhanced L-β-aminoisobutyric acid (L-BAIBA) signalling in the hypothalamus. Red and blue arrows indicate activation and inhibition, respectively.
Figure 2. Proposed hypothesis about the mechanism of clozapine-induced metabolic complication of weight gain associated with enhanced L-β-aminoisobutyric acid (L-BAIBA) signalling in the hypothalamus. Red and blue arrows indicate activation and inhibition, respectively.
Biomolecules 13 01288 g002
Figure 3. Formation and metabolism of β-aminoisobutyric acid (BAIBA) enantiomer.
Figure 3. Formation and metabolism of β-aminoisobutyric acid (BAIBA) enantiomer.
Biomolecules 13 01288 g003
Figure 4. Our proposed hypothesis for the target of action of L-BAIBA on glutamatergic transmission in the thalamocortical pathway. RTN: reticular thalamic nucleus, MDTN: mediodorsal thalamic nucleus, and FPC: prefrontal cortex.
Figure 4. Our proposed hypothesis for the target of action of L-BAIBA on glutamatergic transmission in the thalamocortical pathway. RTN: reticular thalamic nucleus, MDTN: mediodorsal thalamic nucleus, and FPC: prefrontal cortex.
Biomolecules 13 01288 g004
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Fukuyama, K.; Motomura, E.; Okada, M. A Novel Gliotransmitter, L-β-Aminoisobutyric Acid, Contributes to Pathophysiology of Clinical Efficacies and Adverse Reactions of Clozapine. Biomolecules 2023, 13, 1288. https://doi.org/10.3390/biom13091288

AMA Style

Fukuyama K, Motomura E, Okada M. A Novel Gliotransmitter, L-β-Aminoisobutyric Acid, Contributes to Pathophysiology of Clinical Efficacies and Adverse Reactions of Clozapine. Biomolecules. 2023; 13(9):1288. https://doi.org/10.3390/biom13091288

Chicago/Turabian Style

Fukuyama, Kouji, Eishi Motomura, and Motohiro Okada. 2023. "A Novel Gliotransmitter, L-β-Aminoisobutyric Acid, Contributes to Pathophysiology of Clinical Efficacies and Adverse Reactions of Clozapine" Biomolecules 13, no. 9: 1288. https://doi.org/10.3390/biom13091288

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

Article Metrics

Back to TopTop