Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 3747 2022-12-06 19:16:48 |
2 update references and layout Meta information modification 3747 2022-12-07 02:49:08 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Nasyrova, R.F.;  Shnayder, N.A.;  Khasanova, A.K. D-Serine and D-Aspartate in the Pathogenesis of Schizophrenia. Encyclopedia. Available online: https://encyclopedia.pub/entry/38139 (accessed on 02 July 2024).
Nasyrova RF,  Shnayder NA,  Khasanova AK. D-Serine and D-Aspartate in the Pathogenesis of Schizophrenia. Encyclopedia. Available at: https://encyclopedia.pub/entry/38139. Accessed July 02, 2024.
Nasyrova, Regina F., Natalia A. Shnayder, Aiperi K. Khasanova. "D-Serine and D-Aspartate in the Pathogenesis of Schizophrenia" Encyclopedia, https://encyclopedia.pub/entry/38139 (accessed July 02, 2024).
Nasyrova, R.F.,  Shnayder, N.A., & Khasanova, A.K. (2022, December 06). D-Serine and D-Aspartate in the Pathogenesis of Schizophrenia. In Encyclopedia. https://encyclopedia.pub/entry/38139
Nasyrova, Regina F., et al. "D-Serine and D-Aspartate in the Pathogenesis of Schizophrenia." Encyclopedia. Web. 06 December, 2022.
D-Serine and D-Aspartate in the Pathogenesis of Schizophrenia
Edit

Schizophrenia (Sch) is a severe and widespread mental disorder. Antipsychotics (APs) of the first and new generations as the first-line treatment of Sch are not effective in about a third of cases and are also unable to treat negative symptoms and cognitive deficits of schizophrenics. This explains the search for new therapeutic strategies for a disease-modifying therapy for treatment-resistant Sch (TRS). Biological compounds are of great interest to researchers and clinicians, among which D-Serine (D-Ser) and D-Aspartate (D-Asp) are among the promising ones. The Sch glutamate theory suggests that neurotransmission dysfunction caused by glutamate N-methyl-D-aspartate receptors (NMDARs) may represent a primary deficiency in this mental disorder and play an important role in the development of TRS. D-Ser and D-Asp are direct NMDAR agonists and may be involved in modulating the functional activity of dopaminergic neurons.

D-serine D-aspartate disease-modifying therapy treatment-resistant schizophrenia

1. Introduction

Schizophrenia (Sch) is a severe mental disorder that affects approximately 0.5–1% of the population [1]. It includes positive, negative and cognitive symptoms and can lead to significant functional disorders and pronounced social maladaptation of patients [2]. There are several neurochemical hypotheses for the development of Sch: dopaminergic [3], kynurenic [4], glutamatergic [5] and others [6]. The leading hypothesis is dopaminergic, which has formed the basis of the approaches to Sch therapy. Antipsychotics (APs) are the first-line drugs for Sch treatment [7]. However, the issue of achieving a balance between the effectiveness and safety of APs remains open [8]. Other neurochemical hypotheses for the development of Sch continue to be explored as they may provide clues to discover a disease-modifying therapy for mental disorders [9].
The response to APs of the first and new generations (Figure 1) [9] is variable, and it is still difficult to predict whether the therapy will be effective or not [10].
Figure 1. Timeline of antipsychotics (APs) approved by Food and Drug Administration (FDA) [9][11].
About 20–30% of patients with Sch do not have an adequate response ≥2 in terms of dose and duration of AP treatment [12]. This is the clinical definition of treatment-resistant Sch (TRS). As is known, TRS is a serious condition with associated clinical, social and medical expenses and consequences [13]. The definition of TRS has been revised several times. The first definition of TRS was proposed by Kane et al. [14]; this definition is based on the lack of response to Aps, using the example of clozapine. Most new definitions of TRS include failure to respond to at least two consecutive APs courses. In most cases, one of the two APs must be atypical, and have an adequate dose and duration of treatment (≥6 weeks). An adequate dose of an AP in the most recent report is defined as a daily dose equivalent to chlorpromazine ≥ 400 mg [15][16][17]. The absence of a therapeutic response to APs was indicated as a relative change in the evaluation scales (≥20% decrease in the scale of positive and negative symptoms of Sch) [17]. Thus, most patients with TRS may never achieve functional recovery [18], which leads to an increased burden of illness for the patient, family and the Healthcare System [19] (Figure 2).
Figure 2. Burden of treatment-resistant schizophrenia (TRS) for the patient, family and the Healthcare System.
Two types of TRS are known: (1) a type of resistance to APs that is already present at the onset of the disease; (2) the second type of resistance to APs, which develops later during the progression of Sch and/or after a period of successful therapeutic response to APs [20][21].
Treatment methods of Sch that include both typical (the first generation) and atypical (new generations) APs act primarily as brain dopaminergic receptor antagonists. Although these APs are highly effective in treating positive symptoms, their efficacy is limited in patients with persistent negative symptoms or cognitive impairment in patients with Sch [22]. Therefore, the development of a disease-modifying therapy for Sch [23], resistant to APs, is relevant.
Associative genetic studies and genome-wide associative studies have identified over a hundred candidate genes as molecular biomarkers of TRS risk with modest effect [24][25][26]. Interestingly, some of these candidate genes have proteins involved in glutamatergic transmission, especially with the N-methyl-D-aspartate receptors (NMDARs) [27].
So, NMDARs are ligand-dependent ionotropic glutamate receptors, consisting of four subunits [28]. Three families of NMDAR subunits are known: GluN1, GluN2 (subtypes GluN2A, GluN2B, GluN2C and GluN2D) and GluN3 (subtypes GluN3A and GluN3B). At the same time, NMDAR is formed by two GluN1 subunits and either two GluN2 subunits, or a combination of GluN2 and GluN3 subunits, which form a channel (pore) [29]. GluN1 subunits have a glycine co-agonist or D-Serine (D-Ser) recognition sites, while GluN2 subunits have glutamate co-agonist recognition sites. In addition, NMDARs are at rest blocked by Mg2+ ions, which close the Ca2+ channel; NMDAR opens when three things happen simultaneously: (1) glutamate binds to its site on NMDAR; (2) glycine or D-Ser bind to their sites on NMDAR; and (3) depolarization of the neuron membrane occurs. At the same time, depolarization of the cell membrane removes Mg2+ ions from the channel (pore), and the binding of co-agonists Glyc and glutamate provides a voltage-dependent influx of Na+ and Ca2+ ions and outflow of K+ ions, causing postsynaptic effects of glutamate neurotransmission (Figure 3) [30].
Figure 3. Scheme of the N-methyl-D-aspartate receptor (NMDR) functioning. Note: D-Asp—D-aspartate; D-Ser—D-serine; Glyc—glycine.
NMDARs are located predominantly postsynaptically, but can also be located extrasynaptically. Activation of synaptic NMDARs usually promotes the survival of synapses and neurons, while excessive activation of extrasynaptic NMDARs by excess glutamate (“Glutamate shock”) can have a neurotoxic effect and cause neuronal death [31].
D-Aspartate (D-Asp) is a direct NMDAR agonist that excites both metabotropic glutamate receptors and dopaminergic neurons in the brain [32]. In addition, D-Asp is indirectly involved in the initiation and progression of neurodegenerative processes [33] and plays an important role in the neuroplasticity, physiology and morphology of neuronal dendrites, along with regulation of gray matter volume and brain activity [34][35].
D-Ser and D-Asp are D-amino acids [36], and like most amino acids, they have a chiral carbon center, which allows the formation of two stereoisomers. These stereoisomers are mirror images of each other. Similar amino acids have both a left-handed (L) and a right-handed (D) enantiomer (Figure 4) [36].
Figure 4. Scheme of L and D isomers of serine (a) and aspartate (b).
L-amino acids are used in the human body as building blocks of proteins and intermediate products in biochemical processes [37].
Various studies in animal models and humans have demonstrated that D-amino acids, in particular D-Ser and D-Asp (Table 1) [38], are able to modulate various NMDAR-dependent processes, including synaptic plasticity, brain development, cognition and aging brain [39]. Dysfunctional NMDAR activity is associated with the etiology and pathophysiology of a wide range of psychiatric and neurological disorders, including Sch [40]. For example, according to one of the main hypotheses of Sch, glutamate activity in NMDARs is insufficient due to disturbances in the formation of glutamate NMDA synapses during brain development, while this hypothesis not only does not reject, but also confirms, the dopamine hypothesis of Sch [41].
Table 1. Functions of D-aspartate and D-serine in the central nervous system.
This explains the growing number of studies on the role of D-Ser and D-Asp in the pathogenesis of Sch, TRS and disease-modifying therapy for TRS.

2. D-Aspartate

2.1. The Biological Role of D-Aspartate

D-Asp is an endogenous D-amino acid with the molecular formula C4H7NO4. Its average molecular weight is 133.1027 g/mol. D-Asp is located in the cytoplasm of neurons, extracellularly, and in peroxisomes. Laboratory methods detect D-Asp in the blood (21.0 ± 5.0 µM for male, 20.0 ± 5.0 µM for female), cerebrospinal fluid (0–1 µM), feces and urine (<1.13 µmol/mmol creatinine) [42].
Free D-Asp in the human body is determined in the central nervous system (CNS) and the human endocrine system [43]. Endogenous D-Asp is synthesized as a result of racemization of L-Asp in the CNS and endocrine tissue [44]; degradation of dietary protein; and microbial synthesis in the intestine [45].
Exogenous D-Asp enters the human body with food. It is found in vegetables (spinach, beets, iceberg lettuce, avocado, tomato and pepper), some mushrooms (shiitake, oyster mushrooms and chanterelles), fruits and berries (goji, black and red currants, gooseberries, elderberries, grapes and strawberries), some herbs (oregano, spearmint, peppermint, sweet basil, parsley, dill, watercress, rosemary, sorrel, dandelion and fenugreek), fish and nuts [42].
Nervous and endocrine tissues contain the enzymatic systems necessary to modulate D-Asp homeostasis, since they can synthesize and degrade this amino acid. Endogenous racemase activity D-Asp is involved in the biosynthesis of D-Asp from L-Asp, while D-Asp oxidase, a peroxisomal flavoprotein, metabolizes D-Asp to oxaloacetate, NH3 and H2O2 [46].
The level of D-Asp in the CNS increases significantly during fetal development and decreases at birth. Conversely, the level of this amino acid in the endocrine tissue is low during the prenatal period and gradually increases after birth [47] (Figure 5).
Figure 5. Change in D-aspartate balance between brain and endocrine tissue in the human ontogenesis.
Outside the CNS, the D-Asp function is known to regulate the synthesis and secretion of several hormones in endocrine and neuroendocrine tissues [48]. It induces the release of prolactin from the anterior pituitary gland, modulates the production of neurosteroids (oxytocin and vasopressin) in the posterior pituitary gland, and suppresses the secretion of melatonin in the pineal gland. In addition, D-Asp regulates the synthesis and release of testosterone by releasing gonadotropin-releasing hormone (GnRH) in the hypothalamus and luteinizing hormone (LH) in the pituitary gland [46]. In addition, D-Asp can promote animal and human reproduction, also directly activating the proliferation of spermatogonia and improving sperm quality [48].
Functionally, D-Asp corresponds to many, if not all, definitions of a classical neurotransmitter: (1) biosynthesis, degradation, uptake and release of the molecule occur in presynaptic neurons; and (2) D-Asp triggers a response in postsynaptic neurons after it is released from vesicles [49]. A high concentration of D-Asp was first detected in the brain of Octopus vulgaris Lam in 1977 [50]. Later, D-Asp was found in human nervous and endocrine tissues [51][52]. The level of D-Asp during ontogenesis has pronounced regional differences in the CNS, which suggests that D-Asp plays an important role in the development of the CNS. In the brain of rats at the stage of embryonic development, D-Asp is registered near the hindbrain and then spreads to the forebrain. D-Asp was first detected in the body of neurons of the outer layer of the nerve epithelium and then in axons as a distinct axonal layer was formed. These data support the notion that D-Asp is involved in the differentiation of brain neurons [53].
Intense D-Asp immunoreactivity was observed in the cortical plate and subventricular zone of the brain of rats in the early postnatal period. At the same time, D-Asp immunoreactivity decreases to an almost undetectable level in adult rats [54]. In all brain regions studied at all ages, D-Asp immunoreactivity was limited to neurons but not to glia. At the same time, immunohistochemical studies demonstrated D-Asp staining both in the bodies of neurons and in the pathways of neuronal migration. In addition, D-Asp is synthesized in hippocampal and prefrontal cortex neurons, which play an important role in the development of Sch [55].
D-Asp excites dopaminergic neurons and stimulates NMDARs and metabotropic glutamate receptors, preventing neuronal degeneration [32]. D-Asp oxidase regulates the homeostasis of the glutamatergic system when the level of D-Asp in the CNS changes and, thus, suppresses the initiation and progression of neurodegenerative processes [33].
D-Asp binds to the L-glutamate region of ionotropic NMDARs [56][57]. D-Asp in the mouse brain triggers intrinsic currents that counteract competitive and non-competitive NMDAR blockers, including D-2-amino-5-phosphonovaleric acid (D-AP5) and (+)-5-methyl-10,11-dihydro-5H-dibenzo(a,d)cycloheptene-5,10-imine maleate (MK801), ketamine and phencyclidine [58][59]. Residual D-Asp-dependent intrinsic currents persisted even after high doses of these NMDAR antagonists [58][60]. D-Asp also inhibits kainate-induced currents of the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid-type glutamate receptor (AMPAR) in hippocampal neurons and stimulates the metabotropic glutamate receptor 5 (mGluR5) [61][62][63].
D-Asp application triggers intrinsic currents in dopaminergic neurons in the hippocampus, striatum, spinal cord, and substantia nigra compacta by activating NMDAR and, to a lesser extent, AMPA and mGluR5 receptors [59][64].
Studies of primary cultures of neurons and synaptosomal drugs have demonstrated that D-Asp is stored in secretory organelles and released from axon terminals via vesicular exocytosis processes mediated by Ca2+ ions [65][66], or probably by spontaneous release or replacement carriers L-glutamate [67]. D-Asp efficiently crosses the blood–brain barrier (BBB) and is found in nanomolar concentrations in the extracellular space of the prefrontal cortex [63][68]. The level of D-Asp decreases in the CNS depending on the age of the person and is temporarily induced after high stimulation of the flow of K+ ions [69]. It is likely that intracellular D-Asp uptake may depend on the L-glutamate/L-Asp transport systems that recognize L-glutamate and both Asp enantiomers. In vitro studies have shown that D- and L-Asp are recognized and transported by glutamate transporter homolog from Thermococcus kodakarensis (Glt Tk) in the same manner and with comparable affinity [46].
To study the level of endogenous D-Asp, chromatographic methods are used in combination with the enzymatic cleavage of D-Asp. High-performance liquid chromatography (HPLC) and capillary electrophoresis (CE) are used to quantify D-Asp because they can separate chiral amino acids. This allows measurements of each enantiomer [70].

2.2. The Role of D-Aspartate in the Pathogenesis of Schizophrenia

The glutamate theory of Sch occurrence suggests that neurotransmission dysfunction caused by glutamate NMDARs may represent a primary deficit in this mental disorder and the development of TRS [71]. Decreased NMDARs function is considered a key change in the pathophysiology of TRS [10]. Two post-mortem studies have shown a significant decrease in D-Asp levels in prefrontal cortex neurons in Sch patients associated with increased D-Asp oxidase expression [16] or increased D-Asp oxidase enzymatic activity [72]. Sch patients showed altered NMDAR expression [73].
NMDAR agonists may enhance the therapeutic activity of APs in Sch patients, and glutamatergic agents improve negative symptoms of this disease [74].
Conversely, NMDAR antagonists may exacerbate Sch symptoms, and NMDARs hypofunction causes psychosis. NMDAR antagonists modulate dopaminergic activity in nucleus accumbens and different subregions of the prefrontal cortex and this modulation is related to positive symptoms, negative symptoms and cognitive deficits, respectively [75][76]. D-Asp has an effect on synaptic plasticity [34]; its condensate has been found in synaptic vesicles of axon terminals in the developing brain, further supporting the suggestion that D-Asp is an important neurotransmitter involved in CNS development [77].
A number of preclinical studies have demonstrated that D-Asp has an effect on several NMDAR-dependent phenotypes associated with Sch. The D-Asp oxidase gene knockout mice treated with D-Asp demonstrated that long-term elevation of D-Asp levels significantly reduced the deficit in neuronal pre-pulse inhibition induced by psychotomimetic drugs (amphetamine and MK-801) [78]. A single dose of phencyclidine, which simulates the symptoms of Sch in humans and animal models of Sch, reduced motor hyperactivity, ataxia and impairment of pre-pulse inhibition in D-Asp oxidase knockout mice. NMDAR antagonists impair social interaction in rodents an animal model reflecting social withdrawal [10][79]. Elevated D-Asp levels in D-Asp oxidase knockout mice counteract phencyclidine-induced dysfunctional activation of the cortico-limbic-thalamic pathways [79].
It was previously suggested that cortico-hippocampal disconnection in the CNS appears in Sch [80]. In this regard, an increased length of dendrites and a density of spinal cord neurons, as well as a greater functional cortical-hippocampal connectivity in the brains of D-Asp oxidase knockout mice were revealed [79]. An increased functional connection between the hippocampus and the cortex was also found in the rat brain after intragastric administration of D-Asp [81].

3. D-Serine

3.1. Biological Role of D-Serine

D-Ser is an endogenous D-amino acid with the molecular formula C3H7NO3. Its average molecular weight is 105.0926 g/mol. D-Ser is located extracellularly in relation to the neuron. Laboratory methods were found in blood (2.28 ± 0.59 µM), saliva and urine (0.0021–10.1 µmol/mmol creatinine) [82].
Free D-Ser in the human body is determined in the CNS, adipose tissue, epidermis, intestines, kidneys, lungs, pancreas, placenta, platelets, fibroblasts, skeletal muscles, spleen and testicles [82]. Endogenous D-Ser is synthesized by serine racemase (SR), a pyridoxal-5′-phosphate (PLP)-dependent bifunctional type II fold enzyme, from L-Ser [83].
In humans, D-Ser is mainly cleaved by D-amino acid oxidase (DAO). The level of D-Ser in the tissues of the human body is tightly regulated by achieving a balance between its synthesis, degradation, absorption and/or transport. As with most amino acids, when a meal containing Ser is ingested, the molecule is extracted in the small intestine and absorbed into the bloodstream. It crosses the BBB, and enters neurons, where it is metabolized into glycine and many other molecules. Thus, the amount of Ser in cells is regulated by these metabolic processes. If too little is ingested, more Ser is converted from various sources. When too much is ingested, only a portion is converted to glycine, while the rest is metabolized to folate and many other proteins [83].
The level of D-Ser in the CNS remains high during fetal development and postnatal life, even though its content in the brains of adolescents and aged individuals was about half that of fetuses [84] (Figure 6). Researchers did not find data on changes in the level of this amino acid during ontogenesis in endocrine tissues. However, levels of D-serine in the endocrine system are much lower than those in the CNS, and the physiological role of D-serine in the endocrine systems remains unclear [43].
Figure 6. Change in D-serine in the human brain during ontogenesis. Notes: insufficiently studied in endocrine tissue.
Exogenous D-Ser enters the human body with food. It is contained in berries (black raisin, green plum, wampee, goji, hawthorn, lantern fruit, cape gooseberry, mulberry, gooseberry, etc.), fruits (pitaya, lichee, mango, pineapple, guava, banana, coconut, apple and pear), beans, vegetables (olive, avocado, tomato and eggplant), etc. [82].
D-Ser is a potent NMDAR co-agonist, and its role in the brain is of great scientific and clinical interest. D-Ser is present in glia (mainly astrocytes) and CNS neurons. It is considered a glial transmitter [65] and as a neurotransmitter [85]. However, the role of glia and neurons in the therapeutic response to D-Ser is debatable [85]. Volosker et al. [85] suggested that astrocytes synthesize L-Ser, which is then transported to neurons for conversion into D-Ser.
NMDAR activation requires not only glutamate, but also a co-agonist. For many years, glycine was thought to be a co-agonist, and the site where it acts on NMDAR is called the glycine binding site. The regional distribution of D-Ser is more similar to that of NMDRs than that of glycine, and has a stronger affinity than glycine for the glycine binding site on the NR1 subunit of the NMDAR [71]. This indicates that D-Ser may be more important in neurotransmission via NMDARs.
The absence of D-Ser is one of the important factors in the reduction in long-term potentiation and cognition [86]. A decrease in D-Ser reduces the development of long-term potentiation, which is involved in memory consolidation [87]. Elevated D-Ser may improve recognition and memory [88].
The hypofunction of NMDARs has been described in patients with Sch and in an animal model of Sch; therefore, D-Ser is of great interest as the main co-agonist of NMDARs in the forebrain and hippocampus [89]. Glycine and D-Ser seem to act on different populations of NMDARs: D-Ser on synaptic receptors, and glycine on extrasynaptic receptors [32]. Probably, synaptic NMDARs have a neuroprotective effect, and extrasynaptic NMDARs can promote neurodegeneration [90]. An animal model of Sch shows that the basal activity of NMDARs, as well as the branching and density of spikes on neuronal dendrites, are reduced in mice with a knockout of the SR gene encoding SR, an enzyme that converts L-Ser to D-Ser. Long-term administration of D-Ser changed the expression of a protein associated with the neuronal cytoskeleton and caused a partial recovery of dendritic anomalies in the same model [49]. Fujita et al. [91] reported that young and adolescent rodents that were prenatally exposed to the activation of maternal immunity showed reduced expression of NMDAR subunits of hippocampal neurons, which led to the initiation of cognitive impairment in adulthood. In addition, Fujita et al. [91] demonstrated that the addition of D-Ser to drinking water led to a reduction in cognitive deficits in an animal model of Sch. Spatial and reverse memory deficits in Sprague-Dawley rats treated with the NMDAR antagonist phencyclidine can be eliminated by the administration of D-Ser [92].

3.2. The Role of D-Serine in the Development of Schizophrenia

Preclinical studies in an animal model of Sch have demonstrated that a decrease in the level of D-Ser in the CNS due to a decrease in SR activity can cause Sch symptoms, including stereotypy, cognitive impairment, impaired prepulse inhibition (a measure of sensorimotor gating), persistent latent inhibition (a measure of inhibition of learning and cognitive flexibility) and a lack of social interaction [38][93][94].
A decrease in D-Ser levels in cerebrospinal fluid and peripheral blood has been reported in patients with Sch [95]. MacKay et al. [93] reported that the level of D-Ser in the blood of patients with Sch is lower than in the control group, which has also been shown by other researchers [96]. In addition, blood levels of D-Ser and the ratio of D-/L-Ser enantiomers were significantly increased in Sch patients with a good therapeutic response to clozapine (second-generation AP) [68].
Basal D-Ser levels are reduced in Sch patients, potentially due to genetic variation in the genes encoding SR and DAO, which are involved in D-Ser synthesis and degradation, respectively [71]. Significant reductions in D-Ser levels have been found in cerebrospinal fluid in naïve patients with Sch [96]. Balu et al. [97] treated mice with a knockout of the SR gene with D-Ser 300 mg/kg day 1, 150 mg/kg days 2–21 subcutaneously. Restoration of neuroplasticity, increase in BDNF protein expression and reversing the freezing deficit were demonstrated.
Decreased activation of the NMDAR co-agonist site may be seen in TRS, and an increase in D-Ser may reduce the risk and severity of TRS by reversing NMDAR dysfunction [93].
It has been hypothesized that D-Ser may prevent the development of TRS. Pathological and anatomical studies revealed anomalies in the expression of enzymes that modulate D-Ser. Thus, pathoanatomical studies have demonstrated an increase in the expression and activity of DAO in the cerebellum of patients with Sch [90]. Patients with Sch also had higher levels of DAO mRNA in hippocampal neurons [98] and increased DAO activity in cortical neurons [99]. The clinical significance of high levels of DAO in the cerebellum in patients with Sch and TRS is not yet known. However, cerebellar dysfunction in patients with Sch may manifest as mild neurological symptoms or impaired motor function and cognition [100].
Anomalous SR expression in hippocampal and cerebral cortex neurons in patients with Sch [101] has been shown. Although the results of other authors were contradictory and indicated a local increase in the level of SR [61], measures of SR activity in the brains of patients with Sch may be informative in determining whether D-Ser synthesis is altered in this psychiatric disorder.

References

  1. Charlson, F.J.; Ferrari, A.J.; Santomauro, D.F.; Diminic, S.; Stockings, E.; Scott, J.G.; McGrath, J.J.; Whiteford, H.A. Global epidemiology and burden of schizophrenia: Findings from the global burden of disease study 2016. Schizophr. Bull. 2018, 44, 1195–1203.
  2. Vita, A.; Minelli, A.; Barlati, S.; Deste, G.; Giacopuzzi, E.; Valsecchi, P.; Turrina, C.; Gennarelli, M. Treatment-resistant schizophrenia: Genetic and neuroimaging correlates. Front. Pharmacol. 2019, 10, 402.
  3. Howes, O.D.; Shatalina, E. Integrating the neurodevelopmental and dopamine hypotheses of schizophrenia and the role of cortical excitation-inhibition balance. Biol. Psychiatry 2022, 92, 501–513.
  4. Plitman, E.; Iwata, Y.; Caravaggio, F.; Nakajima, S.; Chung, J.K.; Gerretsen, P.; Kim, J.; Takeuchi, H.; Chakravarty, M.M.; Remington, G.; et al. Kynurenic acid in schizophrenia: A systematic review and meta-analysis. Schizophr. Bull. 2017, 43, 764–777.
  5. Uno, Y.; Coyle, J.T. Glutamate hypothesis in schizophrenia. Psychiatry Clin. Neurosci. 2019, 73, 204–215.
  6. Adell, A. Brain NMDA receptors in schizophrenia and depression. Biomolecules 2020, 10, 947.
  7. Seeman, P.; Kapur, S. Schizophrenia: More dopamine, more D2 receptors. Proc. Natl. Acad. Sci. USA 2000, 97, 7673–7675.
  8. Nasyrova, R.F.; Neznanov, N.G. Clinical Psychopharmacogenetics, 1st ed.; DEAN Publishing House: Saint Petersburg, Russia, 2019; pp. 93–174. (In Russian)
  9. Shnayder, N.A.; Khasanova, A.K.; Strelnik, A.I.; Al-Zamil, M.; Otmakhov, A.P.; Neznanov, N.G.; Shipulin, G.A.; Petrova, M.M.; Garganeeva, N.P.; Nasyrova, R.F. Cytokine imbalance as a biomarker of treatment-resistant schizophrenia. Int. J. Mol. Sci. 2022, 23, 11324.
  10. de Bartolomeis, A.; Errico, F.; Aceto, G.; Tomasetti, C.; Usiello, A.; Iasevoli, F. D-aspartate dysregulation in Ddo (−/−) mice modulates phencyclidine-induced gene expression changes of postsynaptic density molecules in cortex and striatum. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2015, 62, 35–43.
  11. : FDA-Approved Drugs. Available online: https://www.accessdata.fda.gov/scripts/cder/daf/index.cfm (accessed on 20 September 2022).
  12. Wagner, E.; Kane, J.M.; Correll, C.U.; Howes, O.; Siskind, D.; Honer, W.G.; Lee, J.; Falkai, P.; Schneider-Axmann, T.; Hasan, A.; et al. Clozapine combination and augmentation strategies in patients with schizophrenia -recommendations from an international expert survey among the treatment response and resistance in psychosis (TRRIP) working group. Schizophr. Bull. 2020, 46, 1459–1470.
  13. Polese, D.; Fornaro, M.; Palermo, M.; De Luca, V.; de Bartolomeis, A. Treatment-resistant to antipsychotics: A resistance to everything? psychotherapy in treatment-resistant schizophrenia and nonaffective psychosis: A 25-year systematic review and exploratory meta-analysis. Front. Psychiatry 2019, 10, 210.
  14. 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.
  15. Ajnakina, O.; Horsdal, H.T.; Lally, J.; MacCabe, J.H.; Murray, R.M.; Gasse, C.; Wimberley, T. Validation of an algorithm-based definition of treatment resistance in patients with schizophrenia. Schizophr. Res. 2018, 197, 294–297.
  16. Elkis, H.; Buckley, P.F. Treatment-Resistant Schizophrenia. Psychiatr. Clin. N. Am. 2016, 39, 239–265.
  17. Remington, G.; Addington, D.; Honer, W.; Ismail, Z.; Raedler, T.; Teehan, M. Guidelines for the pharmacotherapy of schizophrenia in adults. Can. J. Psychiatry 2017, 62, 604–616.
  18. Silverstein, S.M.; Bellack, A.S. A scientific agenda for the concept of recovery as it applies to schizophrenia. Clin. Psychol. Rev. 2008, 28, 1108–1124.
  19. Kennedy, J.L.; Altar, C.A.; Taylor, D.L.; Degtiar, I.; Hornberger, J.C. The social and economic burden of treatment-resistant schizophrenia: A systematic literature review. Int. Clin. Psychopharmacol. 2014, 29, 63–76.
  20. 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.
  21. Sheitman, B.B.; Lieberman, J.A. The natural history and pathophysiology of treatment resistant schizophrenia. J. Psychiatr. Res. 1998, 32, 143–150.
  22. Stępnicki, P.; Kondej, M.; Kaczor, A.A. Current concepts and treatments of schizophrenia. Molecules 2018, 23, 2087.
  23. Bendikov, I.; Nadri, C.; Amar, S.; Panizzutti, R.; De Miranda, J.; Wolosker, H.; Agam, G. A CSF and postmortem brain study of D-serine metabolic parameters in schizophrenia. Schizophr. Res. 2007, 90, 41–51.
  24. Teo, C.; Zai, C.; Borlido, C.; Tomasetti, C.; Strauss, J.; Shinkai, T.; Le Foll, B.; Wong, A.; Kennedy, J.L.; De Luca, V. Analysis of treatment-resistant schizophrenia and 384 markers from candidate genes. Pharm. Genom. 2012, 22, 807–811.
  25. Shen, L.; Lv, X.; Huang, H.; Li, M.; Huai, C.; Wu, X.; Wu, H.; Ma, J.; Chen, L.; Wang, T.; et al. Genome-wide analysis of DNA methylation in 106 schizophrenia family trios in Han Chinese. eBioMedicine 2021, 72, 103609.
  26. Wagh, V.V.; Vyas, P.; Agrawal, S.; Pachpor, T.A.; Paralikar, V.; Khare, S.P. Peripheral blood-based gene expression studies in schizophrenia: A systematic review. Front. Genet. 2021, 12, 736483.
  27. Schizophrenia Working Group of the Psychiatric Genomics Consortium. Biological insights from 108 schizophrenia-associated genetic loci. Nature 2014, 511, 421–427.
  28. Glasgow, N.G.; Siegler Retchless, B.; Johnson, J.W. Molecular bases of NMDA receptor subtype-dependent properties. J. Physiol. 2015, 593, 83–95.
  29. Vieira, M.; Yong, X.; Roche, K.W.; Anggono, V. Regulation of NMDA glutamate receptor functions by the GluN2 subunits. J. Neurochem. 2020, 154, 121–143.
  30. Radulovic, J.; Ren, L.Y.; Gao, C. N-Methyl D-aspartate receptor subunit signaling in fear extinction. Psychopharmacology 2019, 236, 239–250.
  31. Hardingham, G.E.; Fukunaga, Y.; Bading, H. Extrasynaptic NMDARs oppose synaptic NMDARs by triggering CREB shut-off and cell death pathways. Nat. Neurosci. 2002, 5, 405–414.
  32. Krashia, P.; Ledonne, A.; Nobili, A.; Cordella, A.; Errico, F.; Usiello, A.; D′Amelio, M.; Mercuri, N.B.; Guatteo, E.; Carunchio, I. Persistent elevation of D-Aspartate enhances NMDA receptor-mediated responses in mouse substantia nigra pars compacta dopamine neurons. Neuropharmacology 2016, 103, 69–78.
  33. Cristino, L.; Luongo, L.; Squillace, M.; Paolone, G.; Mango, D.; Piccinin, S.; Zianni, E.; Imperatore, R.; Iannotta, M.; Longo, F.; et al. d-Aspartate oxidase influences glutamatergic system homeostasis in mammalian brain. Neurobiol. Aging 2015, 36, 1890–1902.
  34. Errico, F.; Mothet, J.P.; Usiello, A. D-aspartate: An endogenous NMDA receptor agonist enriched in the developing brain with potential involvement in schizophrenia. J. Pharm. Biomed. Anal. 2015, 116, 7–17.
  35. Errico, F.; Nisticò, R.; Di Giorgio, A.; Squillace, M.; Vitucci, D.; Galbusera, A.; Piccinin, S.; Mango, D.; Fazio, L.; Middei, S.; et al. Free D-aspartate regulates neuronal dendritic morphology, synaptic plasticity, gray matter volume and brain activity in mammals. Transl. Psychiatry 2014, 4, e417.
  36. Abdulbagi, M.; Wang, L.; Siddig, O.; Di, B.; Li, B. D-amino acids and d-amino acid-containing peptides: Potential disease biomarkers and therapeutic targets? Biomolecules 2021, 11, 1716.
  37. Biomolecules, Science: Amino Acids: Building Blocks of Proteins. Available online: https://conductscience.com/amino-acids-building-blocks-of-proteins/ (accessed on 20 September 2022).
  38. de Bartolomeis, A.; Vellucci, L.; Austin, M.C.; De Simone, G.; Barone, A. Rational and translational implications of d-amino acids for treatment-resistant schizophrenia: From neurobiology to the clinics. Biomolecules 2022, 12, 909.
  39. Billard, J.M. D-Amino acids in brain neurotransmission and synaptic plasticity. Amino Acids 2012, 43, 1851–1860.
  40. Balu, D.T. The NMDA receptor and schizophrenia: From pathophysiology to treatment. Adv. Pharmacol. 2016, 76, 351–382.
  41. Jiménez-Sánchez, L.; Campa, L.; Auberson, Y.P.; Adell, A. The role of GluN2A and GluN2B subunits on the effects of NMDA receptor antagonists in modeling schizophrenia and treating refractory depression. Neuropsychopharmacology 2014, 39, 2673–2680.
  42. Metabocard for D-Aspartic Acid (HMDB0006483). Human Metabolome Database. Available online: https://hmdb.ca/metabolites/HMDB0006483 (accessed on 20 September 2022).
  43. Kiriyama, Y.; Nochi, H. D-amino acids in the nervous and endocrine systems. Scientifica 2016, 2016, 6494621.
  44. Liang, R.; Robb, F.T.; Onstott, T.C. Aspartic acid racemization and repair in the survival and recovery of hyperthermophiles after prolonged starvation at high temperature. FEMS Microbiol. Ecol. 2021, 97, fiab112.
  45. Bastings, J.; van Eijk, H.M.; Olde Damink, S.W.; Rensen, S.S. D-amino acids in health and disease: A focus on cancer. Nutrients 2019, 11, 2205.
  46. Usiello, A.; Di Fiore, M.M.; De Rosa, A.; Falvo, S.; Errico, F.; Santillo, A.; Nuzzo, T.; Chieffi Baccari, G. New evidence on the role of d-aspartate metabolism in regulating brain and endocrine system physiology: From preclinical observations to clinical applications. Int. J. Mol. Sci. 2020, 21, 8718.
  47. Errico, F.; Napolitano, F.; Nisticò, R.; Usiello, A. New insights on the role of free d-aspartate in the mammalian brain. Amino Acids 2012, 43, 1861–1871.
  48. Di Fiore, M.M.; Santillo, A.; Chieffi Baccari, G. Current knowledge of d-aspartate in glandular tissues. Amino Acids 2014, 46, 1805–1818.
  49. Li, Y.; Han, H.; Yin, J.; Li, T.; Yin, Y. Role of D-aspartate on biosynthesis, racemization, and potential functions: A mini-review. Anim. Nutr. 2018, 4, 311–315.
  50. D′Aniello, A.; Guiditta, A. Identification of d-aspartic acid in the brain of octopus vulgaris lam. J. Neurochem. 1977, 29, 1053–1057.
  51. D′Aniello, G.; Ronsini, S.; Guida, F.; Spinelli, P.; D′Aniello, A. Occurrence of D-aspartic acid in human seminal plasma and spermatozoa: Possible role in reproduction. Fertil. Steril. 2005, 84, 1444–1449.
  52. D′Aniello, G.; Grieco, N.; Di Filippo, M.A.; Cappiello, F.; Topo, E.; D′Aniello, E.; Ronsini, S. Reproductive implication of D-aspartic acid in human pre-ovulatory follicular fluid. Hum. Reprod. 2007, 22, 3178–3183.
  53. Ota, N.; Shi, T.; Sweedler, J.V. D-Aspartate acts as a signaling molecule in nervous and neuroendocrine systems. Amino Acids 2012, 43, 1873–1886.
  54. Wolosker, H.; D′Aniello, A.; Snyder, S.H. D-aspartate disposition in neuronal and endocrine tissues: Ontogeny, biosynthesis and release. Neuroscience 2000, 100, 183–189.
  55. Hons, J.; Zirko, R.; Ulrychova, M.; Cermakova, E.; Doubek, P.; Libiger, J. Glycine serum level in schizophrenia: Relation to negative symptoms. Psychiatry Res. 2010, 176, 103–108.
  56. Singh, S.P.; Singh, V. Meta-analysis of the efficacy of adjunctive NMDA receptor modulators in chronic schizophrenia. CNS Drugs 2011, 25, 859–885.
  57. Sumiyoshi, T.; Anil, A.E.; Jin, D.; Jayathilake, K.; Lee, M.; Meltzer, H.Y. Plasma glycine and serine levels in schizophrenia compared to normal controls and major depression: Relation to negative symptoms. Int. J. Neuropsychopharmacol. 2004, 7, 1–8.
  58. 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.
  59. Tsai, G.E.; Yang, P.; Chung, L.C.; Tsai, I.C.; Tsai, C.W.; Coyle, J.T. D-serine added to clozapine for the treatment of schizophrenia. Am. J. Psychiatry 1999, 156, 1822–1825.
  60. Tsai, G.E.; Lin, P.Y. Strategies to enhance N-methyl-D-aspartate receptor-mediated neurotransmission in schizophrenia, a critical review and meta-analysis. Curr. Pharm. Des. 2010, 16, 522–537.
  61. Verrall, L.; Walker, M.; Rawlings, N.; Benzel, I.; Kew, J.N.; Harrison, P.J.; Burnet, P.W. D-amino acid oxidase and serine racemase in human brain: Normal distribution and altered expression in schizophrenia. Eur. J. Neurosci. 2007, 26, 1657–1669.
  62. Gong, X.Q.; Frandsen, A.; Lu, W.Y.; Wan, Y.; Zabek, R.L.; Pickering, D.S.; Bai, D. D-aspartate and NMDA, but not L-aspartate, block AMPA receptors in rat hippocampal neurons. Br. J. Pharm. 2005, 145, 449–459.
  63. Errico, F.; Nuzzo, T.; Carella, M.; Bertolino, A.; Usiello, A. The emerging role of altered d-aspartate metabolism in schizophrenia: New insights from preclinical models and human studies. Front. Psychiatry 2018, 9, 559.
  64. Weiser, M.; Heresco-Levy, U.; Davidson, M.; Javitt, D.C.; Werbeloff, N.; Gershon, A.A.; Abramovich, Y.; Amital, D.; Doron, A.; Konas, S.; et al. A multicenter, add-on randomized controlled trial of low-dose d-serine for negative and cognitive symptoms of schizophrenia. J. Clin. Psychiatry 2012, 73, e728–e734.
  65. Kantrowitz, J.T.; Malhotra, A.K.; Cornblatt, B.; Silipo, G.; Balla, A.; Suckow, R.F.; D′Souza, C.; Saksa, J.; Woods, S.W.; Javitt, D.C. High dose D-serine in the treatment of schizophrenia. Schizophr. Res. 2010, 121, 125–130.
  66. Wolosker, H.; Sheth, K.N.; Takahashi, M.; Mothet, J.P.; Brady, R.O.; Ferris, C.D., Jr.; Snyder, S.H. Purification of serine racemase: Biosynthesis of the neuromodulator D-serine. Proc. Natl. Acad. Sci. USA 1999, 96, 721–725.
  67. Yamada, K.; Ohnishi, T.; Hashimoto, K.; Ohba, H.; Iwayama-Shigeno, Y.; Toyoshima, M.; Okuno, A.; Takao, H.; Toyota, T.; Minabe, Y.; et al. Identification of multiple serine racemase (SRR) mRNA isoforms and genetic analyses of SRR and DAO in schizophrenia and D-serine levels. Biol. Psychiatry 2005, 57, 1493–1503.
  68. Yamamori, H.; Hashimoto, R.; Fujita, Y.; Numata, S.; Yasuda, Y.; Fujimoto, M.; Ohi, K.; Umeda-Yano, S.; Ito, A.; Ohmori, T.; et al. Changes in plasma D-serine, L-serine, and glycine levels in treatment-resistant schizophrenia before and after clozapine treatment. Neurosci. Lett. 2014, 582, 93–98.
  69. Assisi, L.; Botte, V.; D′Aniello, A.; Di Fiore, M.M. Enhancement of aromatase activity by D-aspartic acid in the ovary of the lizard Podarcis s. sicula. Reproduction 2001, 121, 803–808.
  70. Bi, C.; Zheng, X.; Azaria, S.; Beeram, S.; Li, Z.; Hage, D.S. Chromatographic studies of protein-based chiral separations. Separations 2016, 3, 27.
  71. Kantrowitz, J.T.; Epstein, M.L.; Lee, M.; Lehrfeld, N.; Nolan, K.A.; Shope, C.; Petkova, E.; Silipo, G.; Javitt, D.C. Improvement in mismatch negativity generation during d-serine treatment in schizophrenia: Correlation with symptoms. Schizophr. Res. 2018, 191, 70–79.
  72. Dunlop, D.S.; Neidle, A.; McHale, D.; Dunlop, D.M.; Lajtha, A. The presence of free D-aspartic acid in rodents and man. Biochem. Biophys. Res. Commun. 1986, 141, 27–32.
  73. Beneyto, M.; Kristiansen, L.V.; Oni-Orisan, A.; McCullumsmith, R.E.; Meador-Woodruff, J.H. Abnormal glutamate receptor expression in the medial temporal lobe in schizophrenia and mood disorders. Neuropsychopharmacology 2007, 32, 1888–1902.
  74. Choi, K.H.; Wykes, T.; Kurtz, M.M. Adjunctive pharmacotherapy for cognitive deficits in schizophrenia: Meta-analytical investigation of efficacy. Br. J. Psychiatry 2013, 203, 172–178.
  75. Olney, J.W.; Farber, N.B. Glutamate receptor dysfunction and schizophrenia. Arch. Gen. Psychiatry 1995, 52, 998–1007.
  76. Wu, Q.; Huang, J.; Wu, R. Drugs Based on NMDAR Hypofunction Hypothesis in Schizophrenia. Front. Neurosci. 2021, 15, 641047.
  77. D’Aniello, A. D-Aspartic acid: An endogenous amino acid with an important neuroendocrine role. Brain Res. Rev. 2007, 53, 215–234.
  78. Errico, F.; Rossi, S.; Napolitano, F.; Catuogno, V.; Topo, E.; Fisone, G.; D′Aniello, A.; Centonze, D.; Usiello, A. D-aspartate prevents corticostriatal long-term depression and attenuates schizophrenia-like symptoms induced by amphetamine and MK-801. J. Neurosci. 2008, 28, 10404–10414.
  79. Errico, F.; D′Argenio, V.; Sforazzini, F.; Iasevoli, F.; Squillace, M.; Guerri, G.; Napolitano, F.; Angrisano, T.; Di Maio, A.; Keller, S.; et al. A role for D-aspartate oxidase in schizophrenia and in schizophrenia-related symptoms induced by phencyclidine in mice. Transl. Psychiatry 2015, 5, e512.
  80. Zhou, Y.; Shu, N.; Liu, Y.; Song, M.; Hao, Y.; Liu, H.; Yu, C.; Liu, Z.; Jiang, T. Altered resting-state functional connectivity and anatomical connectivity of hippocampus in schizophrenia. Schizophr. Res. 2008, 100, 120–132.
  81. Kitamura, A.; Hojo, Y.; Ikeda, M.; Karakawa, S.; Kuwahara, T.; Kim, J.; Soma, M.; Kawato, S.; Tsurugizawa, T. Ingested d-aspartate facilitates the functional connectivity and modifies dendritic spine morphology in rat hippocampus. Cereb. Cortex 2019, 29, 2499–2508.
  82. Metabocard for D-Serine (HMDB0003406). Human Metabolome Database. Available online: https://hmdb.ca/metabolites/HMDB0003406 (accessed on 20 September 2022).
  83. Ito, T.; Hamauchi, N.; Hagi, T.; Morohashi, N.; Hemmi, H.; Sato, Y.G.; Saito, T.; Yoshimura, T. D-serine metabolism and its importance in development of dictyostelium discoideum. Front. Microbiol. 2018, 9, 784.
  84. Hashimoto, A.; Kumashiro, S.; Nishikawa, T.; Oka, T.; Takahashi, K.; Mito, T.; Takashima, S.; Doi, N.; Mizutani, Y.; Yamazaki, T. Embryonic development and postnatal changes in free D-aspartate and D-serine in the human prefrontal cortex. J. Neurochem. 1993, 61, 348–351.
  85. Kantrowitz, J.T.; Woods, S.W.; Petkova, E.; Cornblatt, B.; Corcoran, C.M.; Chen, H.; Silipo, G.; Javitt, D.C. D-serine for the treatment of negative symptoms in individuals at clinical high risk of schizophrenia: A pilot, double-blind, placebo-controlled, randomised parallel group mechanistic proof-of-concept trial. Lancet Psychiatry 2015, 2, 403–412.
  86. Yang, S.; Qiao, H.; Wen, L.; Zhou, W.; Zhang, Y. D-serine enhances impaired long-term potentiation in CA1 subfield of hippocampal slices from aged senescence-accelerated mouse prone/8. Neurosci. Lett. 2005, 379, 7–12.
  87. Yang, Y.; Ge, W.; Chen, Y.; Zhang, Z.; Shen, W.; Wu, C.; Poo, M.; Duan, S. Contribution of astrocytes to hippocampal long-term potentiation through release of D-serine. Proc. Natl. Acad. Sci. USA 2003, 100, 15194–15199.
  88. Shimazaki, T.; Kaku, A.; Chaki, S. D-Serine and a glycine transporter-1 inhibitor enhance social memory in rats. Psychopharmacology 2010, 209, 263–270.
  89. Durrant, A.R.; Heresco-Levy, U. D-Serine in neuropsychiatric disorders: New advances. Adv. Psychiatry 2014, 2014, 1–16.
  90. Krystal, J.H.; Karper, L.P.; Seibyl, J.P.; Freeman, G.K.; Delaney, R.; Bremner, J.D.; Heninger, G.R.; Bowers, M.B.; Charney, D.S., Jr. Subanesthetic effects of the noncompetitive NMDA antagonist, ketamine, in humans. Psychotomimetic, perceptual, cognitive, and neuroendocrine responses. Arch. Gen. Psychiatry 1994, 51, 199–214.
  91. Fujita, Y.; Ishima, T.; Hashimoto, K. Supplementation with D-serine prevents the onset of cognitive deficits in adult offspring after maternal immune activation. Sci. Rep. 2016, 6, 37261.
  92. Andersen, J.; Pouzet, B. Spatial Memory Deficits Induced by Perinatal Treatment of Rats with PCP and Reversal Effect of D-Serine. Neuropsychopharmacology 2004, 29, 1080–1090.
  93. MacKay, M.B.; Kravtsenyuk, M.; Thomas, R.; Mitchell, N.D.; Dursun, S.M.; Baker, G.B. D-serine: Potential therapeutic agent and/or biomarker in schizophrenia and depression? Front. Psychiatry 2019, 10, 25.
  94. Taniguchi, K.; Sawamura, H.; Ikeda, Y.; Tsuji, A.; Kitagishi, Y.; Matsuda, S. D-amino acids as a biomarker in schizophrenia. Diseases 2022, 10, 9.
  95. Hons, J.; Zirko, R.; Vasatova, M.; Doubek, P.; Klimova, B.; Masopust, J.; Valis, M.; Kuca, K. Impairment of executive functions associated with lower d-serine serum levels in patients with schizophrenia. Front. Psychiatry 2021, 12, 514579.
  96. Hashimoto, K.; Fukushima, T.; Shimizu, E.; Komatsu, N.; Watanabe, H.; Shinoda, N.; Nakazato, M.; Kumakiri, C.; Okada, S.; Hasegawa, H.; et al. Decreased serum levels of D-serine in patients with schizophrenia: Evidence in support of the N-methyl-D-aspartate receptor hypofunction hypothesis of schizophrenia. Arch. Gen. Psychiatry 2003, 60, 572–576.
  97. Balu, D.T.; Li, Y.; Puhl, M.D.; Benneyworth, M.A.; Basu, A.C.; Takagi, S.; Bolshakov, V.Y.; Coyle, J.T. Multiple risk pathways for schizophrenia converge in serine racemase knockout mice, a mouse model of NMDA receptor hypofunction. Proc. Natl. Acad. Sci. USA 2013, 110, E2400–E2409.
  98. Habl, G.; Zink, M.; Petroianu, G.; Bauer, M.; Schneider-Axmann, T.; von Wilmsdorff, M.; Falkai, P.; Henn, F.A.; Schmitt, A. Increased D-amino acid oxidase expression in the bilateral hippocampal CA4 of schizophrenic patients: A post-mortem study. J. Neural Transm. 2009, 116, 1657–1665.
  99. Burnet, P.W.; Eastwood, S.L.; Bristow, G.C.; Godlewska, B.R.; Sikka, P.; Walker, M.; Harrison, P.J. D-amino acid oxidase activity and expression are increased in schizophrenia. Mol. Psychiatry 2008, 13, 658–660.
  100. Phillips, J.R.; Hewedi, D.H.; Eissa, A.M.; Moustafa, A.A. The cerebellum and psychiatric disorders. Front. Public Health 2015, 3, 66.
  101. Labrie, V.; Fukumura, R.; Rastogi, A.; Fick, L.J.; Wang, W.; Boutros, P.C.; Kennedy, J.L.; Semeralul, M.O.; Lee, F.H.; Baker, G.B.; et al. Serine racemase is associated with schizophrenia susceptibility in humans and in a mouse model. Hum. Mol. Genet. 2009, 18, 3227–3243.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , ,
View Times: 646
Entry Collection: Biopharmaceuticals Technology
Revisions: 2 times (View History)
Update Date: 07 Dec 2022
1000/1000
Video Production Service