Abstract
Psychedelic compounds are being increasingly explored as a potential therapeutic option for treating several psychiatric conditions, despite relatively little being known about their mechanism of action. One such possible mechanism, DNA methylation, is a process of epigenetic regulation that changes gene expression via chemical modification of nitrogenous bases. DNA methylation has been implicated in the pathophysiology of several psychiatric conditions, including schizophrenia (SZ) and major depressive disorder (MDD). In this review, we propose alterations to DNA methylation as a converging model for the therapeutic effects of psychedelic compounds, highlighting the N-methyl D-aspartate receptor (NMDAR), a crucial mediator of synaptic plasticity with known dysfunction in both diseases, as an example and anchoring point. We review the established evidence relating aberrant DNA methylation to NMDAR dysfunction in SZ and MDD and provide a model asserting that psychedelic substances may act through an epigenetic mechanism to provide therapeutic effects in the context of these disorders.
INTRODUCTION
Epigenetic modifications, such as DNA methylation and histone modification, describe a collection of genetic changes that alter gene expression without changing the base pair order of the genome. Given their ability to respond to environmental stimuli with lasting effects on gene expression, the field of epigenetics is particularly attractive to researchers studying diseases with environmental influences, and in recent years, epigenetic mechanisms have been increasingly implicated in the pathophysiology of several psychiatric diseases [1, 2]. Amongst the psychiatric diseases to demonstrate epigenetic changes are schizophrenia (SZ) and major depressive disorder (MDD). Despite the differing symptom profiles of these two disorders, evidence supports the role of epigenetic modification, and specifically DNA methylation, in the development, presentation, and progression of both SZ and MDD.
DNA methylation is the process of adding a methyl group to cytosine residues in the DNA, which affects the ability of DNA transcription machinery to transcribe the DNA, ultimately resulting in a change in gene expression. As discussed below, DNA methylation induces different effects on gene transcription and subsequent protein expression levels based on the location of methylation along the gene. While DNA methylation of the promoter likely leads to decreased expression [3–5], DNA methylation of the gene body may lead to increased expression levels [6–9]. This versatility allows DNA methylation to be considered for various pathophysiological phenomena and across a diverse set of psychiatric diseases. One of the many phenomena potentially explained by DNA methylation is the dysfunction of the N-methyl D-aspartate receptor (NMDAR), which has been implicated in the pathophysiology of both SZ and MDD, albeit in seemingly opposite directions.
In SZ, NMDAR hypofunction is one of the leading theories of disease etiology and is believed to contribute to cortical hyperactivity and disruptions in other neurotransmitter systems [10]. In MDD, in contrast, the theory of NMDAR hyperactivity has been posited following the observed antidepressant effects associated with various NMDAR antagonists, most notably ketamine [11]. Ketamine’s mechanism of therapeutic action has not yet been fully elucidated, however, and some research has suggested that the antidepressant effects may be independent of ketamine’s NMDAR inhibition [12]. The rapid onset antidepressant action of ketamine and other NMDAR antagonists lends credibility to the notion of NMDAR dysfunction in MDD, however, the exact nature of this dysfunction involves complex synaptic and circuit mechanisms [13, 14]. NMDAR function is dependent not only on the number and location of the receptors, but also on receptor subunit composition. DNA methylation has the potential to explain NMDAR dysfunction in both SZ and MDD by resulting in altered levels of specific NMDAR subunits, as well as global decreases or increases in NMDAR activity.
In examining the NMDAR dysfunction in MDD, it is difficult not to discuss the recent rise in preclinical research and clinical trials examining the antidepressant actions of psychedelic compounds in the treatment of MDD [15]. Despite having different receptor binding targets, drugs from various classes of psychedelic compounds share many subjective and antidepressant effects, suggesting a common downstream effector. A commonly proposed mechanism for this therapeutic effect is a change in synaptic plasticity [16], a process which involves modulation of NMDAR activity. Thus, there is a connection between the NDMAR dysfunction seen in psychiatric illnesses, including MDD and SZ, and the potential mechanism of action underlying the antidepressant effects of psychedelic compounds. We argue here that this relationship can be explained by a convergent mechanism: epigenetic modification, namely DNA methylation. Additional investigation of the epigenetic effects of psychedelic compounds is needed to fully uncover their therapeutic potential, which may extend to other psychiatric diseases and symptom groups, potentially including the negative symptoms of SZ.
EPIGENETIC MODIFICATIONS: DNA METHYLATION
Epigenetic modifications are alterations to the genome that do not affect the base pair order of the DNA and can occur transiently in response to environmental cues. Since first proposed in 1942 [17], the field of epigenetics has expanded dramatically. Epigenetic modifications have been implicated in the pathophysiology of numerous developmental processes and disease states, including in neuropsychiatric disorders. One of the most well-characterized processes of epigenetic modification, DNA methylation, is a process wherein DNA methyltransferases (DNMTs) transfer a methyl group from S-adenyl methionine (SAM) onto a cytosine residue, forming 5mC [18]. Here, we provide a brief overview of the process of DNA methylation, followed by a review of the role of DNA methylation in the pathophysiology of SZ and MDD.
There are several known DNMT proteins, including DNMT1, DNMT3A, DNMT3B, and DNMT3L (Fig. 1). DNMT1 preserves DNA methylation patterns during DNA replication and cell division using hemimethylated DNA as a template [19]. DNMT3A and DNMT3B function to methylate DNA de novo, i.e., without requiring a hemimethylated template [20]. DNMT3L does not have catalytic activity, but it associates with the de novo methyltransferases (DNMT3A and DNMT3B) to stimulate activity [21]. Though not exclusively, the vast majority of DNA methylation occurs at cytosines that are immediately adjacent to a guanine, so-called CpG sites [22, 23], and the majority of human CpG sites are methylated [24, 25].
Fig. 1. DNA methylation by DNA methyltransferases.

A DNMT1 preserves DNA methylation patterns during DNA replication and cell division. B DNMT3A and DNMT3B perform de novo DNA methylation; DNMT3L acts in association with these DNA methyltransferases to stimulate activity. C Increased DNA methylation in the promoter region interferes with gene transcription and results in decreased gene expression. In contrast, increased DNA methylation in the gene body does not interfere with gene transcription and may result in increased gene expression.
There exist stretches of the genome approximately 1 kb in length with marked increases in CpG frequency, known as CpG islands (CGIs) [26]. Despite the high concentration of CpG sites, most CpG islands are unmethylated, potentially due to the tendency toward conversion of 5mC to thymine via deamination, resulting in a germline loss of CpGs outside of CpG islands [27]. Most promoters exist within CpG islands and are unlikely to be methylated [28]. Approximately one third of intragenic CGIs in the human brain are methylated, although the function of this tissue-specific intragenic methylation is currently unknown [29]. Methyl-CpG binding proteins (MeCPs), so called DNA methylation “readers,” recognize methylation marks at CpG dinucleotides to alter gene expression [30]. For example, MeCP2 can associate with co-repressor complexes to induce transcription repression [31, 32], and mutations to the MeCP2 gene (MECP2) have been directly associated with the neurological disorder Rett syndrome [33], highlighting the correlation between epigenetic regulation and neurological function.
DNA methylation alters the transcription of a particular genomic sequence, however, the location of the methylation along the gene appears to dictate the nature of this alteration (Fig. 1). DNA methylation in the promoter region of a gene can impair the ability of transcriptional machinery to bind to and transcribe DNA, thus resulting in decreased expression of the gene product (gene repression) [3–5, 34]. However, in actively dividing cells, DNA methylation of the gene body does not interfere with gene transcription, and in fact can result in increased gene expression [6–9]. The three-dimensional chromosomal conformation of the DNA may also be affected by DNA methylation, which could facilitate or inhibit interactions between promoters and enhancers, further influencing genic expression [35]. DNA methylation may also play a role in transcription product splicing, as increased methylation is observed at the boundaries between exons and introns [36].
DNA METHYLATION IN SZ AND MDD
Genome-wide association studies (GWAS) have identified a number of genetic risk loci for both SZ [37] and MDD [38] that fall within genetic regulatory domains, including non-coding regions and promoters, supporting the notion that aberrant regulation of gene expression and other genetic processes may be a significant contributor in the pathophysiology of both diseases. Epigenetic modification, especially DNA methylation, significantly contributes to gene expression regulation, and thus epigenetic aberrations have been investigated as a possible mechanism underlying the development and pathophysiology of SZ and MDD.
One of the most consistent findings in epigenetic investigations of SZ and SZ-relevant phenotypes is the overexpression of DNMT1 [39–42]. Several studies have found that DNMT1 is overexpressed specifically in the GABAergic interneurons, but not pyramidal neurons, of SZ patients [42–44]. DNMT1 functions by moving a methyl group onto cytosine residues, and treatment with methionine, a methyl-group donor, markedly worsens psychotic symptoms in patients with SZ [45]. Administration of methionine to cortical neuron cultures results in methylation of the RELN promoter, as well as subsequent downregulation of reelin and glutamic acid decarboxylase 67 (GAD67) mRNA expression, an effect that was reversed by knockdown of DNMT1 [46]. Thus, administration of methionine likely exacerbates the effects of the already hyperactive DNMTs in SZ by providing an additional substrate.
Several epigenomic studies have found differential DNA methylation patterns in SZ and MDD compared to control samples. SZ- or psychosis-relevant differentially methylated CpGs have been identified in both postmortem prefrontal cortex (PFC) [47] and peripheral blood of living subjects [48, 49]. A recent study identified 66 CpG sites wherein methylation was associated with depression score in monozygotic twins [50]. Additionally, this study identified 19 differentially methylated regions between the depressed and non-depressed pairs, and in the majority, methylation was positively correlated to depression score, indicating a trend of hypermethylation in depression.
Investigators have also utilized animal models and human tissue to interrogate the methylation status of specific genes associated with disease pathology, sometimes yielding mixed results. In human studies, some investigations of DNA methylation in SZ have shown hypermethylation of the RELN promoter [51–54] (though others have failed to detect a statistically significant difference [55, 56]) and hypermethylation of the GAD1 (glutamate decarboxylase 1 gene) promoter in the inferior parietal lobe of postmortem psychotic patients [57]. However, others found hypomethylation of this same gene in repressive chromatin of postmortem SZ tissue [58] and hypomethylation of the membrane-bound catechol-O-methyltransferase (MB-COMT) gene body (COMT), with subsequent downregulation of the protein [59–61]. Consistent with dopaminergic hyperactivity in SZ, there was DNA hypermethylation in the gene body of dopamine receptors D3 (DRD3) [62] and D4 (DRD4) (males only) [63] but hypomethylation in the promoter regions of D2 (DRD2) [64].
Chronic stress is a cornerstone in the study of animal models of depression, and investigations utilizing this model have demonstrated a number of stress-induced epigenetic alterations. A recent meta-analysis showed that chronic unpredictable mild stress (CUMS) is a robust method to elicit anhedonia in animal models [65]. Investigations have shown decreased histone acetylation in the hippocampus of rats exposed to chronic stress [66] and induction of DNMT3A in the nucleus accumbens (NAc) of rats exposed to social defeat stress [67], amongst many others (reviewed in refs. [68, 69]). Furthermore, differential DNA methylation and histone modification of the glial cell-derived neurotrophic factor gene (GDNF) reliably predicted response to stress in two distinct mouse lines [70]. Research has also shown that stress is related to DNA methylation changes in several genes in humans, including a glucocorticoid receptor gene (NR3C1), a serotonin transporter gene (SLC6A4), and BDNF, amongst others (reviewed in ref. [71]). Interestingly, acute inhibition of stress-induced DNA methylation using DNMT inhibitors promotes rapid and sustained antidepressant effects associated with increased BDNF-TrkB-mTOR signaling in the PFC [72].
As outlined above, investigations into the epigenetic modifications in SZ and MDD often yield mixed results. These conflicting findings may result from yet undefined disease subtypes underlying varied pathophysiology. A recent study demonstrated decreased methylation of the oxytocin receptor gene (OXTR) in both recent onset SZ and ultra-high risk for psychosis cohorts, while hypomethylation was associated with increased negative symptoms in both SZ and psychosis women [73]. This supports the notion that aberrations in DNA methylation at distinct loci may contribute to a subtype of SZ with more pronounced negative symptoms. Further, when SZ patients were divided into two subtypes, one with marked difference in methylation status compared to healthy controls and one with methylation patterns that were relatively similar to healthy controls [74], the differential methylation group demonstrated more severe symptomology and degree of cognitive deficit. The potential role of DNA methylation in the generation of different subtypes of SZ should be further explored, with the weighted contribution to other symptom domains considered.
As in SZ, DNA methylation patterns also help to explain the varied symptom presentations associated with depression. In a 2021 study, methylation profiles differed in the peripheral blood of patients with adult-onset versus late-onset depression, and several CpG sites were identified as potential markers of adult-onset depression [75]. While the age of disease onset may be related to different methylation patterns in depression, epigenetic aging on its own does not appear accelerated in the depressed individual [76]. Epigenetic alterations may also be responsible for sex-specific differences in disease manifestation and symptom presentation. In a subchronic variable defeat stress model, female mice appeared more susceptible to developing depression-like symptoms than their male counterparts and demonstrated greater induction of DNMT3A in the NAc [77]. Interestingly, when DNMT3A was upregulated in the NAc of male mice, it increased their susceptibility to developing depression-like symptoms, while DNMT3A knockdown in female mice showed an increase in resilience to the development of depressive-like symptoms.
Presented above are only a handful of the many known aberrations in DNA methylation that have been identified in SZ and MDD, supporting widespread changes in epigenetic regulation in the pathophysiology of both diseases. Additionally, both SZ and MDD demonstrate dysfunction of the NMDAR, and we propose that this dysfunction may be in part mediated by DNA methylation aberrations. Next, we will briefly review the NMDAR, discuss NMDAR dysfunction in SZ and MDD, and finally present the limited research on DNA methylation of the NMDAR subunit genes in both disorders.
NMDARS
The NMDAR is a class of ionotropic glutamate receptors activated by NMDA, in addition to glutamate [78] and glycine [79]. These receptors are indispensable for synaptic plasticity-dependent processes such as learning and memory and are implicated in a number of psychiatric and neurological disorders. Other non-NMDA ionotropic glutamate receptors include AMPA receptors and kainate receptors. The NMDAR contains two glutamate binding sites and two glycine binding sites [80], and is composed of 2 obligatory GluN1 subunits and 2 GluN2 or GluN3 subunits [81–84]. GluN1 can include up to 8 splice variants as a result of posttranscriptional RNA processing [85, 86]. GluN2 includes 4 variants encoded by the GluN2A-D genes (GRIN2A-D) [87, 88], and GluN3 includes 2 variants encoded by the GluN3A-B genes (GRIN3A-B) [89]. The genes for the NMDAR subunits are dispersed throughout the genome, spanning five chromosomes (Fig. 2).
Fig. 2. NMDA receptor subunits.

A Genes for the NMDA receptor subunits are dispersed across five chromosomes – chromosome 9 (GluN1 and GluN3A), chromosome 12 (GluN2B), chromosome 16 (GluN2A), chromosome 17 (GluN2C), and chromosome 19 (GluN2D and GluN3B). B Most NMDA receptors are heterodimeric, composed of two obligatory GluN1 subunits and two identical GluN2 or GluN3 subunits. C Some heterotrimeric NMDA receptors have been identified, which are composed of two GluN1 subunits and either (1) one GluN2A subunit + one GluN2B subunit, (2) one GluN2A subunit + one GluN2C subunit, or (3) one GluN2B subunit + one GluN2D subunit. D The GluN2A subunit is ubiquitously expressed throughout the adult brain, while GluN2B predominates in the forebrain, GluN2C predominates in the cerebellum, and GluN2D predominates in the midbrain. E Throughout development, GluN2B subunits are replaced with GluN2A subunits, in what is known as the GluN2B-to-GluN2A switch.
NMDARs are moved between synaptic and extrasynaptic sites via rapid lateral translocation [90], and the process of NMDAR trafficking is implicated in long-term depression (LTD) and potentiation (LTP). The subunit composition of NMDARs has significant consequences on receptor function and kinetic profile. NMDAR subunit composition also plays a significant role in the relative contribution of NMDARs to the processes of LTD and LTP. Within a few hours following synaptic activation, NMDA receptors, particularly GluN2A-containing receptors, are preferentially recruited to synapses [91, 92] and thus play a more prominent role in LTP induction, partly due to their faster kinetics [93]. In contrast, extrasynaptic NMDARs are predominately GluN2B-containing [94], which are strongly implicated in LTD [95]. Notably, the process of NMDAR trafficking, and thus synaptic plasticity, is influenced by BDNF levels (reviewed in ref. [96]). BDNF has been shown to increase GluN1, GluN2A, and GluN2B mRNA and protein levels in the hippocampus [97], and the process of hippocampal LTP is mediated in part via signaling of the BDNF receptor TrkB [98].
In addition to their differential contributions to LTP/LTD, the subunit composition of NMDARs follows distinct developmental and brain-region specific patterns (Fig. 2). While the GluN1 subunit is ubiquitously expressed in all NDMARs, GluN2 subunit expression patterns are more variable, with GluN2A subunits being widely distributed throughout the adult brain: GluN2B subunits predominate in the forebrain [99, 100], GluN2C subunits in the cerebellum, and GluN2D subunits in the midbrain and interneuron populations [101–104]. Tri-heteromeric NMDARs (GluN1/GluN2A/GluN2B) are concentrated predominately in the hippocampus and cortex [105–107].
GluN2B and GluN2D subunits are expressed in the embryonic brain, and GluN2A expression increases from birth through development. GluN2A subunits increase in proportion to GluN2B throughout development, and this GluN2B-to-GluN2A subunit transition is critical to normal neurological development [102, 108]. Although GluN2B subunits predominate in the cerebellum at birth, they largely disappear from this region during early postnatal development and are replaced by GluN2C subunits, which, during the same timespan, disappear from forebrain structures and are replaced by GluN2B subunits [102]. GluN2D expression decreases dramatically throughout development [102].
Given their critical role in normal and pathophysiological processing, positive and negative modulators of NMDARs have been well-characterized (reviewed in refs. [109, 110]). Positive modulators of NMDAR function include polyamines, arachidonic acid, pregnenolone sulfate, and CIQ. Negative modulators of NMDAR function include ketamine, D-APV, memantine, MK-801, TCN-201, ifenprodil, pregnanolone sulfate, and QNZ46. These positive and negative modulators have been used to investigate the effects of NMDAR hypo- and hyperfunction on behavior, development, and disease states, and have contributed to our understanding of various neuropsychiatric disorders. Below, we will discuss the role of NDMAR dysfunction SZ and MDD, and we will describe the potential role of DNA methylation in this dysfunction.
NMDAR DYSFUNCTION IN SZ
Although the dopamine hypothesis once reigned as the predominate view of neurotransmitter dysfunction in the pathophysiology of SZ, many experts now argue that dysfunction of the NMDAR-mediated glutamate system may be a more apt explanation, at least for early development and progression of SZ. Evidence of NMDAR dysfunction in SZ dates back to the 1990s, when it was discovered that in healthy control subjects, the NMDAR antagonists ketamine and PCP induced SZ-relevant symptoms [111–114], and these same substances exacerbated symptoms in SZ patients [115, 116]. Since then, details of the glutamate hypothesis of SZ, the notion that NMDAR hypofunction is a core tenet of the pathophysiology of SZ, have been extensively researched and reviewed [10, 100, 117–119].
NMDAR dysfunction in SZ is most often characterized as hypofunction of the NMDARs of GABAergic interneurons, resulting in disinhibition of cortical pyramidal neurons (Fig. 3). NMDAR hypofunction in SZ is supported by numerous postmortem examinations of SZ brain tissue, which have revealed aberrations in several NMDAR subunit protein and mRNA levels, though results related to specific subunit levels have been mixed. A meta-analysis showed decreased GluN1 mRNA expression in the PFC of SZ patients [120], whereas GRIN2A was identified as a genetic risk locus for the development of SZ [121, 122]. Decreased activity of the downstream signaling cascades associated with the NMDAR was also found in postmortem examination of the dorsolateral PFC in SZ patients, specifically in the postsynaptic density fraction, despite increased NMDAR density in this same area [123], suggesting NMDAR may be dysfunctional in SZ, rather than reduced in quantity.
Fig. 3. Proposed mechanisms of NMDAR dysfunction in SZ and MDD.

DNA methylation of the promoter and gene body result in different expression profiles, and this may underlie the opposing directions of NMDAR dysfunction in SZ (hypofunction) and MDD (hyperfunction). A DNA methylation of the GRIN2B Promoter in SZ. DNA methylation of the GRIN2B promoter prevents transcription of the GRIN2B gene, resulting in decreased GluN2B expression. This GRIN2B promoter hypermethylation may be partially responsible for the NMDAR hypofunction in SZ, demonstrated in Fig. A.1–3. A.1 NMDA receptors on GABAergic interneurons are hypoactive, preventing the activation of these interneurons. A.2 The interneuron does not release GABA at the synaptic junction between GABAergic interneuron and pyramidal neuron. A.3 The pyramidal neuron is disinhibited, resulting in increased firing and cortical hyperexcitability. B DNA Methylation of the GRIN2A Body in MDD. DNA methylation of the GRIN2A gene body facilitates transcription of the GRIN2A gene, resulting in increased GluN2A expression. This GRIN2A gene body hypermethylation may be partially responsible for the NMDAR hyperfunction proposed in MDD, demonstrated in Fig. B.1–3. B.1 NMDA receptors on GABAergic interneurons are hyperactive, resulting in activation of these interneurons. B.2 The interneuron releases GABA at the synaptic junction between GABAergic interneuron and pyramidal neuron. B.3 The pyramidal neuron is inhibited, resulting in decreased firing and cortical hypoexcitability.
Support for NMDAR hypofunction and subsequent cortical disinhibition in the pathophysiology of SZ has also been demonstrated in numerous animal models of psychosis (Table 1). Electrophysiological studies have revealed a number of changes associated with SZ, perhaps the most notable and consistent of which is the increased gamma-band oscillations observed in SZ patients and SZ-relevant animal models [124]. In the healthy brain, parvalbumin (PV)- and somatostatin (SST)-containing interneurons contribute inhibitory inputs to pyramidal cortical neurons to support the maintenance of gamma oscillations [125]. Decreased PV interneuron activity due to NMDAR hypofunction may result in hyperexcitability of the cortex, thus contributing to the alterations in gamma synchrony typically seen in SZ [126]. The NMDAR antagonist ketamine also increases the power of gamma-band oscillations [127, 128], and in a model of NMDAR hypofunction, serine racemase knockout (SRKO) mice demonstrate impaired investigation-elicited gamma power and increased background gamma power, suggesting a deficit in signal-to-noise ratio in the gamma band, which may contribute to impaired cognitive functioning [129]. Finally, SRKO mice exhibited reduced inhibitory synapses onto CA1 pyramidal neurons, which resulted in an increased E/I balance [130]. Each of these studies provide supporting evidence for the concept of NMDAR hypofunction resulting in disinhibition of cortical pyramidal neurons.
Table 1.
Human and animal studies investigating NMDAR subunit aberrations in SZ and MDD.
| Sample/Population studied | Relevant disease | NMDAR subunit(s)/allele(s) investigated | Methods | Results | Reference |
|---|---|---|---|---|---|
| Cre recombinase mice | SZ | GluN1 (GRIN1) | GRIN1 alleles disrupted in ~50% of cortical and hippocampal interneurons in the second postnatal week | GRIN1-deficient mice demonstrated several SZ-relevant behaviors following social isolation | [200] |
| Cre recombinase mice | SZ | GluN1 (GRIN1) | Amphetamine-induced dopamine changes were investigated in GRIN-1 deficient mice | GRIN1-deficient mice demonstrated dopaminergic disruptions characteristic of SZ - amphetamine increased dopamine in the nucleus accumbens and decreased dopamine in the mPFC | [201] |
| Cre recombinase mice | SZ | GluN1 (GRIN1) | GRIN1 alleles disrupted in ~50% of cortical and hippocampal interneurons in the second postnatal week | GRIN1-deficient mice demonstrated upregulation of glycogen synthase kinase-3 (GSK3) in GABAergic interneurons, and GSK3B inhibition resulted in decreased SZ-relevant phenotypes in these mice. | [202] |
| MK-801 treated rats | SZ | GluN2A (GRIN2A), GluN2B (GRIN2B) | Pharmacologic models of SZ were used to investigate levels of GRIN2A- and GRIN2B-targeting micro RNAs (miRNAs) | Elevated levels of GRIN2A/2B-targeting miRNAs were identified in both SZ-relevant models, and decreased expression levels of GluN2A and GluN2B protein were also seen in both models | [203] |
| Methylazoxy-methanol acetate (MAM) treated rats | SZ | GluN2A (GRIN2A), GluN2B (GRIN2B) | |||
| Postmortem human PFC samples | SZ, MDD | GluN1 isoforms (GRIN1 splice variants) | Postmortem PFC from patients with depression, SZ, or nonpsychiatric healthy controls were assessed for GluN1 splice variant levels | C1 cytosolic segment-containing GluN1 levels were decreased in SZ and increased in MDD | [204] |
| Postmortem human lateral amygdala samples | MDD | GluN1 (GRIN1), GluN2A (GRIN2A) | GluN1 and GluN2A protein levels were measured in postmortem depressed or nonpsychiatric healthy control samples | GluN2A levels were significantly increased in depressed populations; GluN1 levels were not significantly different | [205] |
| Postmortem human locus coeruleus and cerebellum samples | MDD | GluN1 (GRIN1), GluN2C (GRIN2C) | GluN1 and GluN2C protein levels were measured in postmortem depressed or nonpsychiatric healthy control samples | GluN2C levels were significantly increased in the locus coeruleus of depressed subjects; GluN1 levels were not significantly different | [206] |
| Postmortem human locus coeruleus and PFC samples | MDD | All NMDAR subunits (GluN1, GluN2A-D, GluN3A-B) | NMDAR subunit protein levels were measured in postmortem depressed or nonpsychiatric healthy control samples | GluN2B and GluN2C subunits were upregulated in the locus coeruleus, but not the PFC, of depressed subjects | [207] |
| Postmortem human dorsolateral PFC | MDD | GluN1 (GRIN1), GluN2A-D (GluN2A-D) | GluN1 and GluN2A-D levels were increased in female MDD subjects | [208] | |
| Postmortem human PFC samples | MDD | GluN1 (GRIN1), GluN2A (GRIN2A), GluN2B (GRIN2B) | GluN1, GluN2A, and GluN2B protein levels were measured in postmortem depressed or nonpsychiatric healthy control samples | GluN2A and GluN2B levels were reduced in the PFC of depressed subjects; GluN1 levels were not significantly different | [209] |
| Postmortem human medial temporal lobe samples | MDD | GluN1 (GRIN1), GluN2A-D (GluN2A-D) | NMDAR subunit protein levels were measured in postmortem samples from SZ, MDD, or bipolar disorder patients, and from nonpsychiatric healthy controls | GluN2A and GluN2B levels were reduced in the PFC of depressed subjects; GluN1 levels were not significantly different | [210] |
| Postmortem human dorsolateral PFC | MDD, SZ | GluNI (GRIND, GluN2A-D (GluN2A-D) | GluN1 levels were decreased in MDD, SZ, and bipolar disorder; GluN2A levels were decreased in SZ and MDD; and GluN2C levels were decreased in SZ; GluN2B and GluN2D levels were not significantly different | [211] |
The timing of NMDAR hypofunction is critical in the development of SZ or SZ-relevant symptoms, as the age at which NMDAR hypofunction is induced appears to dictate the effects seen and the degree to which SZ-like behaviors are generated. Following administration of the NMDAR antagonist MK-801 on postnatal day 10, mice demonstrated reduced function of PV-containing interneurons, decreased GABA transmission, and decreased levels of GABA-related proteins [50]. Additionally, these mice showed several SZ-relevant behavioral deficits in adolescence. Alvarez et al. investigated the effects of early postnatal NMDAR ablation in cortical and hippocampal interneurons at different developmental stages and found that while cortical hyperactivity occurred in the juvenile mice, the mPFC to hippocampus functional hypoconnectivity was not evident until adolescence [131].
These findings support the sequential contribution of cortical hyperactivity to the later development of SZ-like phenotypes. The GluN2B-to-GluN2A transition in NMDAR composition during development occurs later in the PFC than in other brain regions, as discussed above, and this corresponds to later maturation of this brain region as compared to others [99]. NMDAR hypofunction early in development may exacerbate this delayed transition, and further contribute to the cognitive dysfunction seen in SZ. Interestingly, repeated administration of (R)-ketamine to maternal immune activation-exposed animals during adolescence resulted in a significant reduction in SZ-relevant symptoms in adulthood [132]. This suggests that (R)-ketamine, which contains only the (R) enantiomer of the normally racemic ketamine mixture (discussed below), may have neuroprotective effects when administered during development, and this theory warrants further investigation.
The theory of NMDAR hypofunction in SZ is further supported by the dysfunction of other neurotransmitters seen in SZ, as many of these neurotransmitter systems work synergistically with the glutamatergic system. In our recent review, we argued that the NMDAR hypofunction seen in SZ was not a result of aberrant neuromodulatory transmitters (e.g., dopamine), but rather that NMDAR hypofunction resulted in the subsequent dysfunction of other systems [100]. The reciprocal relationship between NMDAR-mediated neurotransmission and various other neuromodulatory systems is extensive, and studies investigating NMDAR hypofunction have revealed numerous other neurotransmitter system aberrations, including increased dopamine release in the striatum [133], aberrant BDNF signaling in the PFC [134], and increased serotonin levels in the mPFC [135].
NMDAR DYSFUNCTION IN MDD
Support for NMDAR dysfunction in MDD was first evidenced by the antidepressant effects of various NMDAR antagonists. The discovery of this drug class in the treatment of depression came about serendipitously, when D-cycloserine was administered for the treatment of tuberculosis but additionally resulted in elevated mood [136]. This finding was expounded upon by Trullas and Skolnick, who found that NMDAR antagonists, including the non-competitive antagonist MK-801 and the competitive antagonist AP-7, have antidepressant-like effects in animal models of depression [137]. Since these initial findings, NDMAR antagonists have demonstrated significant antidepressant effects in both preclinical and clinical studies. Notably, the non-competitive NMDAR antagonist ketamine has consistently demonstrated rapid, long-lasting antidepressant effects in numerous clinical populations [138–142] (reviewed in refs. [13, 143]).
Ketamine’s antidepressant mechanism of action is currently an area of active research and has not yet been fully elucidated. Much in the same way that NMDAR hypofunction in SZ is theorized to increase cortical activity via preferential blockade of NMDARs on GABAergic (inhibitory) interneurons, resulting in disinhibition of the cortical pyramidal neurons, ketamine results in increased cortical activity despite blocking excitatory (glutamatergic) neurotransmission (Fig. 3) [144, 145]. It is not clear if this NMDAR blockade is responsible for the antidepressant effects of ketamine, as recent studies have demonstrated the potential for an NMDAR-independent mechanism related to ketamine metabolites [12]. Furthermore, as noted above, the antidepressant effects of ketamine outlast the acute NMDAR blockade of the drug [14].
Ketamine consists of the (R) and (S) enantiomers, commonly referred to as arketamine and esketamine, respectively. (S)-ketamine has significantly higher NMDAR binding affinity than (R)-ketamine [146], and the vast majority of clinical trials investigating individual ketamine enantiomers for the treatment of MDD have focused on (S)-ketamine rather than (R)-ketamine. Despite lower NMDAR binding affinity, (R)-ketamine may have more potent antidepressant effects based on preclinical studies [147–149]. Furthermore, (R)-ketamine produces fewer psychosis-like side effects [147] and has lower abuse liability [150] than (S)-ketamine, and thus may be a more tolerable treatment option. Indeed, in rhesus monkeys, esketamine, but not arketamine, decreased dopamine D2/3 receptor binding in the striatum [151], in line with the increased risk of psychosis-like effects of esketamine compared to arketamine. (R)- and (S)-ketamine also produced differential fMRI responses when administered to conscious rats, with (R)-ketamine mimicking the effects elicited following MK-801 administration [152]. This finding suggests that the antidepressant mechanism underlying (R)-ketamine is unique from that underlying (S)-ketamine, and that (R)-ketamine may potentially function in an NMDAR-independent manner. However, despite apparently lacking GluN2B subunit specificity, ketamine’s antidepressant effects are negated in GluN2B-deficient mice, suggesting a GluN2B-specific role in ketamine’s mechanism of action [153].
Given the knowledge that NMDAR antagonism produces antidepressant effects, some researchers have investigated NMDAR hyperactivity as a possible mechanism in the pathophysiology of MDD. Postmortem studies have revealed various aberrations in the expression levels of NMDAR subunits in the brains of patients with MDD [118] (see Table 1).
Several rodent models of depression have also demonstrated stress-related increases in NMDAR subunit expression. These include increased GluN1 expression/mRNA levels in the hippocampus following immobilization stress [154, 155] and increased GluN1 subunit levels in the VTA following varied stress paradigm [156]; increased GluN2A subunit expression in adult rats exposed to maternal separation [157]; increased GRIN1, GRIN2A, and GRIN2B expression in the frontal cortex of BDNF-deficient animals [158]; and increased GluN2A and GluN2B mRNA expression in the hippocampus following chronic restraint stress [159] and chronic corticosterone administration [160]. Of note, chronic stress exposure resulted in increased GluN1 subunit expression, but no significant changes in mRNA levels of GluN2A or GluN2B in rat hippocampus [161]. In animals exposed to chronic unpredictable mild stress, GRIN1 expression was decreased [162]. Chronic stress has also been shown to decrease NMDAR subunit expression (GluN1, GluN2A, and GluN2B) in the PFC, but not the motor cortex, of rats [163].
In summary, these studies of NMDAR subunit aberration in depression, when taken together with studies of antidepressant effects of NMDAR antagonists, suggest that NMDAR hyperactivity may contribute to the symptom profiles seen in MDD. Furthermore, NMDAR subunit dysregulation may contribute to the aberrations in synaptic plasticity seen in MDD, though the exact contributions of NMDAR dysregulation to the pathophysiology of depression are still being elucidated. This concept is further discussed in our section on the therapeutic potential of psychedelics, which may exert their antidepressant functions through NMDAR-mediated changes in LTP and synaptic plasticity (see Fig. 4).
Fig. 4. Psychedelic compounds, mechanisms of action in synaptic plasticity.

A, A.1 Psychedelic compounds include classic psychedelics (psilocybin, LSD), which act as 5HT2A receptor agonists; dissociative psychedelics (ketamine), which act as NMDAR antagonists; entactogens (MDMA), which act as positive modulators of the serotonin and dopamine systems; and atypical psychedelics (ibogaine, DMT), which do not fit into the aforementioned categories. A.2 In addition to their individual receptor binding targets, psychedelic compounds may also bind to the BDNF receptor TrkB. B Despite their varied receptor binding targets, psychedelic compounds may share downstream signaling pathways that ultimately result in changes to, and/or stabilization of, DNA methylation patterns. C DNA methylation patterns result in changes to NMDAR subunit expression, ultimately resulting in increased dendritic spine formation and changes to NMDAR-mediated synaptic plasticity, which may underlie the therapeutic effects of psychedelic compounds.
DNA METHYLATION OF NMDAR SUBUNIT GENES
As outlined above, alterations in DNA methylation patterns and aberrations in epigenetic machinery in SZ and MDD have been established by extensive research, much of which is still ongoing. Given the evidence supporting both epigenetic alterations and NMDAR dysfunction in SZ and MDD, it is reasonable to theorize that this NMDAR dysfunction may result from aberrant epigenetic regulation. Still, oddly, relatively few studies have investigated this specific line of questioning. Therefore, we will now focus on the limited research on DNA methylation changes in SZ and MDD associated explicitly with the NMDAR.
In a recent study, hypomethylation of the GRIN2B promoter was found in peripheral blood samples of SZ patients, and methylation at a particular CpG site was correlated positively to cognitive function [164]. However, in a study investigating the relationship between childhood trauma and methylation status at GRIN1, GRIN2A, and GRIN2B in the peripheral blood of patients with SZ, their siblings, and healthy controls, GRIN1 hypermethylation was found to be correlated to trauma status in siblings but not SZ patients [165]. Rats reared in isolation also demonstrated hypermethylation of GRIN1 in the PFC and of GRIN2B in the hippocampus [166]. Interestingly, subchronic PCP (an NMDAR antagonist) administration results in promoter hypermethylation of the GRIN1 and GRIN2B genes, but not the GRIN2A gene, in rodent models [167].
Still, it is unknown exactly how epigenetic mechanisms may regulate NMDAR subunits. Repressor element 1-silencing transcription factor (REST) contributes to the GluN2B-to-GluN2A switch seen during neurodevelopment via epigenetic regulation, and both REST expression and methylation of H3K27 at the GRIN2B promoter are increased in the methylazoxymethanol (MAM) rodent model of SZ [168]. NMDAR blockade with the competitive antagonist CGP-37849 resulted in aberrant histone acetylation in the mPFC of rodents, suggesting that NMDAR hypofunction and epigenetic alterations could be reciprocal in SZ [169]. Indeed, it is possible that NMDAR hypofunction in early adolescence results in DNA methylation of NDMAR-relevant genes, which perpetuates a state of continued NMDAR hypofunction throughout an individual’s life.
An interesting example of higher-order chromatin relevant for SZ was reported for the GRIN2B gene locus [35, 117, 170]. Specifically, this higher-order chromatin is dynamically regulated by neuronal activity, and multiple loop-bound intronic and intergenic DNA sequences from the GRIN2B transcription start site (TSS) are formed to compete for promoter access to regulate GRIN2B expression [35, 170]. In addition to such promoter-enhancer loops, the GRIN2B promoter interacts with and is counterbalanced by intragenic repressive chromatin embedded in intron sequences downstream from the TSS [170]. Transcriptional regulation at the GRIN2B locus is thus likely engaged with a dynamic and competitive interplay of multiple loop formations involving the GRIN2B promoter, which may be further influenced by methylation status at the promoter and distant regulatory sites [170, 171]. Interestingly, these loop-bound sequences interact with the GRIN2B promoter and harbor single nucleotide polymorphisms (SNP) implicated in working memory and SZ [170].
In a genome-wide methylation study of 94 maltreated and 96 non-traumatized children, GRIN1 was identified as one of three genes that predicted depression [172]. Further, in validating these genes in a mouse model of neglect, methylation of GRIN1, along with DNA-binding protein inhibitor (ID3) and tubulin polymerization-promoting protein (TPPP), significantly predicted depression-like behaviors in the elevated plus maze and forced swim test [173]. GRIN2A hypermethylation has also been identified in the PFC and hippocampus of depressed individuals compared to healthy controls following examination of postmortem brain tissue [174].
These studies support the role of DNA methylation in the differential regulation of GRIN2B gene expression in SZ (Fig. 3). Given the role of GluN2B subunit-containing NMDARs in the induction of LTD, and the known deficits in synaptic plasticity in neuropsychiatric disorders like SZ and MDD, this is particularly interesting. Still, additional studies are needed to understand the role of this differential expression in the pathophysiology of SZ. The differential methylation and subsequent altered expression of the GRIN2B gene in SZ are of particular interest given both the GluN2B-to-GluN2A transition during neurodevelopment and the known dysfunction of the PFC, where the GluN2B subunit predominates, in SZ patients. Additionally, more research is needed to elucidate the role of DNA methylation in the expression regulation of other NMDAR subunits. Of note, recent studies have consistently reported the critical role of the GRIN2A gene in SZ [121, 122]. Whether and how the GRIN2A gene is methylated, and how this process may contribute to the progression of SZ and depression, remain to be determined.
THE THERAPEUTIC POTENTIAL OF PSYCHEDELICS
The recent success of ketamine as a potential treatment for depression has sparked a renewed interest in the use of psychedelic compounds for the treatment of numerous psychiatric disorders. While ketamine may not immediately evoke the essence of a psychedelic compound, given its action as an NMDAR antagonist rather than a serotonin agonist, psychedelic compounds include an array of substances with varying effects and receptor binding targets (see Fig. 4). Psychedelic compounds are generally divided into classes of classic psychedelics, which function as serotonin 2A (5-HT2A) agonists (e.g., psilocybin and LSD); dissociative psychedelics, which function as NMDAR antagonists (e.g., ketamine); entactogens, which function as positive modulators of both the serotonin and dopamine systems (e.g., MDMA); and atypical psychedelics, which do not fit into the above categories (reviewed in ref. [175]). The hallmark psychedelic effects of these compounds generally include profound perceptual and emotional alterations, including euphoria, auditory and visual hallucinations, time distortions, and ego dissolution [176].
Psychedelic compounds have had a complicated history in the medical field, as many early drug experiments were steeped in unethical practices and resulted in a decades-long moratorium on government-funded psychedelic research in the US [175]. In more recent years, psychedelic compounds have again become an appealing area of psychiatric research, as they have shown promise for treating several different psychiatric illnesses, including depression, post-traumatic stress disorder, anxiety, obsessive-compulsive disorder, and substance use disorders (reviewed in ref. [15]). Despite the increase in psychedelic-mediated treatment trials, the mechanism by which these substances elicit their therapeutic effects is still unclear. Given the varied receptor binding targets of psychedelic compounds, a shared downstream effector action is expected, and several theories have been proposed to describe this potential mechanism of psychedelic therapeutic action.
One of the most common changes elicited by psychedelic administration in both animal and human studies is that of increased synaptic plasticity. Both psilocybin [177] and N,N-dimethyltryptamine (DMT) [178] have been shown to increase the density of dendritic spines in the brains of rodents. DMT, LSD, and DOI also increase synaptogenesis in rodents [179]. Synaptic plasticity has been theorized to underlie the increases in cognitive flexibility observed in human studies of psychedelic compounds [180]. In a recent study of psilocybin for the treatment of MDD, improvements in depressive symptoms were correlated with increases in theta power on subject EEG [181]. Exactly how these observations of neural and cognitive plasticity relate to one another, however, is an active area of research.
One proposed mechanism involves binding psychedelic compounds to the BDNF receptor TrkB. In a 2023 study, Moliner and colleagues found that both LSD and psilocin, the active ingredient in psilocybin, bind to TrkB with greater affinity than non-psychedelic antidepressant compounds and that this TrkB binding is necessary for the synaptic plasticity effects seen following psychedelic administration [182]. This discovery is particularly intriguing because psychedelic-induced TrkB signaling does not require serotonin 5-HT2A receptor signaling, suggesting that the plasticity changes associated with psychedelics may be independent of their hallucinogenic properties. Similarly, it was shown that the behavioral and electrophysiological responses to psilocybin are preserved in mice despite the coadministration of ketanserin, a 5-HT2A/2C receptor antagonist [183].
The idea that the therapeutic mechanism of psychedelic action may be independent of serotonin 2A receptor signaling is particularly attractive, as it helps bridge the gap between different psychedelic subclasses. Despite sharing common subjective and therapeutic effects, not all psychedelic compounds share a common binding target. Most notably, ketamine, a potent dissociative psychedelic with known therapeutic benefits in the treatment of MDD, functions as an NMDAR antagonist. The possibility of a shared downstream effector mechanism of psychedelic action despite varied receptor binding targets was recently supported in a series of studies by Nardou and colleagues, in which it was demonstrated that psychedelic compounds across multiple subclasses consistently function to reopen a critical period of social reward learning [184, 185].
First, Nardou and colleagues found that MDMA could reopen a critical period of social reward learning via oxytocin-dependent LTD synaptic plasticity [185]. Given that MDMA is an empathogenic psychedelic, generally resulting in increased sociability and social reward, the authors then sought to discover if this ability to reopen the social reward learning critical period was specific to MDMA or shared across psychedelics. They proceeded to demonstrate that ketamine, LSD, psilocybin, and ibogaine were all able to reopen to social reward learning critical period in rodents following a single dose [184]. They further established that the duration of the critical period reopening was proportional to the duration of subjective effects typically seen in humans. Theorizing that this critical period reopening may be accomplished via downstream effects on gene expression regulation, they investigated mRNA expression levels following psychedelic administration and found differential expression of several genes associated with the extracellular matrix (ECM). Here, we propose that psychedelic compounds may induce alterations in DNA methylation patterns, resulting in altered expression of genes related to synaptic plasticity, including NMDAR subunit genes, and that these changes in DNA methylation contribute to the therapeutic effects of the drugs (Fig. 4).
The concept of drug-induced epigenetic modifications is not new. It has been well-established that cannabinoid exposure is associated with differential DNA methylation of various genes in peripheral blood [186–188], sperm [189, 190], and brain tissue [191, 192]. Furthermore, it has been shown that in animals with parental THC exposure, there is significant GRIN2A hypomethylation in exon 1 and decreased GluN2A gene expression, supporting the notion of drug-induced DNA methylation changes resulting in aberrant gene expression [193]. Differential DNA methylation patterns are also observed in methamphetamine-treated mice compared to control groups [194]. More recently and perhaps most excitingly, widespread DNA methylation changes have been identified in the rodent PFC following repeated administration of the psychedelic compound LSD [195]. It is reasonable to theorize, then, that other substances may also induce changes in DNA methylation, and that these changes may be related to the underlying psychiatric effects of these substances. Here, we discuss briefly the limited research investigating DNA methylation changes associated with psychedelic compounds.
Despite the apparent knowledge gap regarding the mechanism of action underlying ketamine’s antidepressant effects, very little research has been done into the epigenetic modifications induced by ketamine treatment. In the chronic despair mouse model of depression, both acute ketamine treatment and chronic antidepressant (imipramine and fluoxetine) treatment produced significant decreases in DNA methylation of the Homer1 (a scaffolding protein associated with synaptic signaling) promoter [196]. This reduction in DNA methylation levels was a reversal of the promoter hypermethylation and decreased expression seen in the chronic despair model prior to treatment. It was also associated with an increase in Homer1 gene expression following treatment. In addition, ketamine treatment, when combined with fear extinction training, produced significant decreases in BDNF promoter DNA methylation in an inescapable foot shock model [197]. This reduction was again a reversal of observed hypermethylation induced by inescapable foot shock prior to treatment. These two studies not only demonstrate the capacity of ketamine to reduce DNA methylation, but they also offer the possibility that ketamine reverses aberrant methylation in psychiatric disease, potentially underlying ketamine’s mechanism of antidepressant action. In a mouse model of depression, DOI (2,5-dimethyoxy-4-iodoamphetamine) not only significantly reduced depressive-like symptoms, but also resulted in long-lasting epigenomic changes, including gene clusters related to NDMAR function [198].
CONCLUSIONS AND FUTURE DIRECTIONS
Despite the rapid rise of psychedelic medications for the treatment of various psychiatric disorders, and the known aberrations in DNA methylation observed in these disorders, very little research has been conducted to examine a potential link between the antidepressant effects of psychedelic substances and the epigenetic modifying properties of these substances. We propose that alterations in DNA methylation underlie the antidepressant effects of psychedelic compounds, using the NMDAR genes as a reference point. NMDAR dysfunction is implicated in the pathophysiology of depression, via hyperactivity of the GABAergic interneurons and subsequent hypoactivity of the cortex, supported by the antidepressant properties of the NMDAR antagonist ketamine. This NMDAR hyperactivity may partly be caused by aberrations in DNA methylation patterns associated with these genes, and treatment with psychedelic compounds may reverse these aberrations.
Further, we described the NMDAR hypofunction that results in decreased activity of GABAergic interneurons and subsequent hyperactivity of the cortex in SZ. Several lines of research have implicated DNA methylation aberrations in the NMDAR dysfunction in SZ. Many psychedelic compounds have been shown to elicit psychosis-like effects in healthy controls and worsen symptoms in patients with SZ when administered acutely. However, there is limited research investigating the changes in DNA methylation patterns observed following the administration of psychedelic compounds to NMDAR-deficient animal models. Psychedelic compounds may have a stabilizing effect on DNA methylation patterns, which could result in reduced symptoms for some psychiatric concerns beyond their known antidepressant effects. Additionally, it is unclear the mechanism by which psychedelic compounds alter DNA methylation patterns. This should be further investigated, as it could offer therapeutic potential for treating SZ, amongst other psychiatric illnesses, if the mechanism could be separated from the acute psychedelic effects of these compounds. For example, the non-hallucinogenic psychedelic analogue tabernanthalog was recently synthesized and demonstrated potential therapeutic effects in the treatment of substance use and depression-like behaviors in rodent models [199].
The age at which NMDAR dysfunction begins, and the age at which psychedelic compounds are administered, may impact the long-term symptom profile of the individual. As discussed above, the NMDAR subunit preponderance is not only brain region-specific, but changes throughout development, most notably in the so-called GluN2B-to-GluN2A subunit transition that occurs early in development. It is unknown how changes in DNA methylation affect this transition, and how interventions aimed at altering DNA methylation levels may ultimately affect development. Furthermore, NMDAR subunits predominate in different ratios based on the specific cell, and the effects of NDMAR subunit epigenetic modification at a cellular level, e.g., pyramidal neurons vs. GABAergic interneurons, have yet to be fully elucidated. Finally, as medicine generally and psychiatric treatment specifically become increasingly more individualized, it raises the question of disease subtypes and SNP-specific genotypes in the pathophysiology of disease, and the effects of methylation status of these disease-specific SNPs should be further explored for diagnostic and treatment purposes.
ACKNOWLEDGEMENTS
We thank Mr. Jake D. Clarin for his comments on the manuscript. The images were created with BioRender.com. This study was supported by NIH R21MH121836, R21MH129989, R01MH131053, and Pennsylvania Commonwealth 4100085747 (CURE 2016) to WJG.
Footnotes
COMPETING INTERESTS
The authors declare no competing interests.
REFERENCES
- 1.Richetto J, Meyer U. Epigenetic modifications in schizophrenia and related disorders: molecular scars of environmental exposures and source of phenotypic variability. Biol Psychiatry. 2021;89:215–26. [DOI] [PubMed] [Google Scholar]
- 2.Penner-Goeke S, Binder EB. Epigenetics and depression. Dialogues Clin Neurosci. 2019;21:397–405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Levine A, Cantoni GL, Razin A. Inhibition of promoter activity by methylation: possible involvement of protein mediators. Proc Natl Acad Sci USA. 1991;88:6515–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Boyes J, Bird A. Repression of genes by DNA methylation depends on CpG density and promoter strength: evidence for involvement of a methyl-CpG binding protein. Embo J. 1992;11:327–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Hwu WL, Lee YM, Lee SC, Wang TR. In vitro DNA methylation inhibits FMR-1 promoter. Biochem Biophys Res Commun. 1993;193:324–9. [DOI] [PubMed] [Google Scholar]
- 6.Aran D, Toperoff G, Rosenberg M, Hellman A. Replication timing-related and gene body-specific methylation of active human genes. Hum Mol Genet. 2011;20:670–80. [DOI] [PubMed] [Google Scholar]
- 7.Ball MP, Li JB, Gao Y, Lee JH, LeProust EM, Park IH, et al. Targeted and genomescale strategies reveal gene-body methylation signatures in human cells. Nat Biotechnol. 2009;27:361–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Hellman A, Chess A. Gene body-specific methylation on the active X chromosome. Science. 2007;315:1141–3. [DOI] [PubMed] [Google Scholar]
- 9.Rauch TA, Wu X, Zhong X, Riggs AD, Pfeifer GP. A human B cell methylome at 100-base pair resolution. Proc Natl Acad Sci USA. 2009;106:671–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Nakazawa K, Sapkota K. The origin of NMDA receptor hypofunction in schizophrenia. Pharm Ther. 2020;205:107426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Chan SY, Matthews E, Burnet PW. ON or OFF? Modulating the N-Methyl-D-aspartate receptor in major depression. Front Mol Neurosci. 2016;9:169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Zanos P, Moaddel R, Morris PJ, Georgiou P, Fischell J, Elmer GI, et al. NMDAR inhibition-independent antidepressant actions of ketamine metabolites. Nature. 2016;533:481–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Krystal JH, Abdallah CG, Sanacora G, Charney DS, Duman RS. Ketamine: a paradigm shift for depression research and treatment. Neuron. 2019;101:774–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Krystal JH, Kavalali ET, Monteggia LM. Ketamine and rapid antidepressant action: new treatments and novel synaptic signaling mechanisms. Neuropsychopharmacology. 2023. 10.1038/s41386-023-01629-w. Online ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Vollenweider FX, Preller KH. Psychedelic drugs: neurobiology and potential for treatment of psychiatric disorders. Nat Rev Neurosci. 2020;21:611–24. [DOI] [PubMed] [Google Scholar]
- 16.Aleksandrova LR, Phillips AG. Neuroplasticity as a convergent mechanism of ketamine and classical psychedelics. Trends Pharm Sci. 2021;42:929–42. [DOI] [PubMed] [Google Scholar]
- 17.Deichmann U Epigenetics: the origins and evolution of a fashionable topic. Dev Biol. 2016;416:249–54. [DOI] [PubMed] [Google Scholar]
- 18.Kumar S, Cheng X, Klimasauskas S, Mi S, Posfai J, Roberts RJ, et al. The DNA (cytosine-5) methyltransferases. Nucleic Acids Res. 1994;22:1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Yen RW, Vertino PM, Nelkin BD, Yu JJ, el-Deiry W, Cumaraswamy A, et al. Isolation and characterization of the cDNA encoding human DNA methyltransferase. Nucleic Acids Res. 1992;20:2287–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Xie S, Wang Z, Okano M, Nogami M, Li Y, He WW, et al. Cloning, expression and chromosome locations of the human DNMT3 gene family. Gene. 1999;236:87–95. [DOI] [PubMed] [Google Scholar]
- 21.Suetake I, Shinozaki F, Miyagawa J, Takeshima H, Tajima S. DNMT3L stimulates the DNA methylation activity of Dnmt3a and Dnmt3b through a direct interaction. J Biol Chem. 2004;279:27816–23. [DOI] [PubMed] [Google Scholar]
- 22.Grippo P, Iaccarino M, Parisi E, Scarano E. Methylation of DNA in developing sea urchin embryos. J Mol Biol. 1968;36:195–208. [DOI] [PubMed] [Google Scholar]
- 23.Bianchi NO, Vidal-Rioja L, Cleaver JE. Direct visualization of the sites of DNA methylation in human, and mosquito chromosomes. Chromosoma. 1986;94:362–6. [DOI] [PubMed] [Google Scholar]
- 24.Edwards JR, O’Donnell AH, Rollins RA, Peckham HE, Lee C, Milekic MH, et al. Chromatin and sequence features that define the fine and gross structure of genomic methylation patterns. Genome Res. 2010;20:972–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Ehrlich M, Gama-Sosa MA, Huang LH, Midgett RM, Kuo KC, McCune RA, et al. Amount and distribution of 5-methylcytosine in human DNA from different types of tissues of cells. Nucleic Acids Res. 1982;10:2709–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Bird A, Taggart M, Frommer M, Miller OJ, Macleod D. A fraction of the mouse genome that is derived from islands of nonmethylated, CpG-rich DNA. Cell. 1985;40:91–99. [DOI] [PubMed] [Google Scholar]
- 27.Bird AP. DNA methylation and the frequency of CpG in animal DNA. Nucleic Acids Res. 1980;8:1499–504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Ioshikhes IP, Zhang MQ. Large-scale human promoter mapping using CpG islands. Nat Genet. 2000;26:61–63. [DOI] [PubMed] [Google Scholar]
- 29.Maunakea AK, Nagarajan RP, Bilenky M, Ballinger TJ, D’Souza C, Fouse SD, et al. Conserved role of intragenic DNA methylation in regulating alternative promoters. Nature. 2010;466:253–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Meehan RR, Lewis JD, McKay S, Kleiner EL, Bird AP. Identification of a mam-malian protein that binds specifically to DNA containing methylated CpGs. Cell. 1989;58:499–507. [DOI] [PubMed] [Google Scholar]
- 31.Kokura K, Kaul SC, Wadhwa R, Nomura T, Khan MM, Shinagawa T, et al. The Ski protein family is required for MeCP2-mediated transcriptional repression. J Biol Chem. 2001;276:34115–21. [DOI] [PubMed] [Google Scholar]
- 32.Jones PL, Veenstra GJ, Wade PA, Vermaak D, Kass SU, Landsberger N, et al. Methylated DNA and MeCP2 recruit histone deacetylase to repress transcription. Nat Genet. 1998;19:187–91. [DOI] [PubMed] [Google Scholar]
- 33.Kyle SM, Vashi N, Justice MJ. Rett syndrome: a neurological disorder with metabolic components. Open Biol. 2018;8:170216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Mohn F, Weber M, Rebhan M, Roloff TC, Richter J, Stadler MB, et al. Lineage-specific polycomb targets and de novo DNA methylation define restriction and potential of neuronal progenitors. Mol Cell. 2008;30:755–66. [DOI] [PubMed] [Google Scholar]
- 35.Rajarajan P, Jiang Y, Kassim BS, Akbarian S. Chromosomal conformations and epigenomic regulation in schizophrenia. Prog Mol Biol Transl Sci. 2018;157:21–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Laurent L, Wong E, Li G, Huynh T, Tsirigos A, Ong CT, et al. Dynamic changes in the human methylome during differentiation. Genome Res. 2010;20:320–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Roussos P, Mitchell AC, Voloudakis G, Fullard JF, Pothula VM, Tsang J, et al. A role for noncoding variation in schizophrenia. Cell Rep. 2014;9:1417–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Wray NR, Ripke S, Mattheisen M, Trzaskowski M, Byrne EM, Abdellaoui A, et al. Genome-wide association analyses identify 44 risk variants and refine the genetic architecture of major depression. Nat Genet. 2018;50:668–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Zhubi A, Veldic M, Puri NV, Kadriu B, Caruncho H, Loza I, et al. An upregulation of DNA-methyltransferase 1 and 3a expressed in telencephalic GABAergic neurons of schizophrenia patients is also detected in peripheral blood lymphocytes. Schizophr Res. 2009;111:115–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Dong E, Ruzicka WB, Grayson DR, Guidotti A. DNA-methyltransferase1 (DNMT1) binding to CpG rich GABAergic and BDNF promoters is increased in the brain of schizophrenia and bipolar disorder patients. Schizophr Res. 2015;167:35–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Veldic M, Caruncho HJ, Liu WS, Davis J, Satta R, Grayson DR, et al. DNA-methyltransferase 1 mRNA is selectively overexpressed in telencephalic GABAergic interneurons of schizophrenia brains. Proc Natl Acad Sci USA. 2004;101:348–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Veldic M, Guidotti A, Maloku E, Davis JM, Costa E. In psychosis, cortical interneurons overexpress DNA-methyltransferase 1. Proc Natl Acad Sci USA. 2005;102:2152–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Ruzicka WB, Zhubi A, Veldic M, Grayson DR, Costa E, Guidotti A. Selective epigenetic alteration of layer I GABAergic neurons isolated from prefrontal cortex of schizophrenia patients using laser-assisted microdissection. Mol Psychiatry. 2007;12:385–97. [DOI] [PubMed] [Google Scholar]
- 44.Veldic M, Kadriu B, Maloku E, Agis-Balboa RC, Guidotti A, Davis JM, et al. Epigenetic mechanisms expressed in basal ganglia GABAergic neurons differentiate schizophrenia from bipolar disorder. Schizophr Res. 2007;91:51–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Antun FT, Burnett GB, Cooper AJ, Daly RJ, Smythies JR, Zealley AK. The effects of L-methionine (without MAOI) in schizophrenia. J Psychiatr Res. 1971;8:63–71. [DOI] [PubMed] [Google Scholar]
- 46.Noh JS, Sharma RP, Veldic M, Salvacion AA, Jia X, Chen Y, et al. DNA methyltransferase 1 regulates reelin mRNA expression in mouse primary cortical cultures. Proc Natl Acad Sci USA. 2005;102:1749–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Jaffe AE, Gao Y, Deep-Soboslay A, Tao R, Hyde TM, Weinberger DR, et al. Mapping DNA methylation across development, genotype and schizophrenia in the human frontal cortex. Nat Neurosci. 2016;19:40–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Hannon E, Dempster EL, Mansell G, Burrage J, Bass N, Bohlken MM, et al. DNA methylation meta-analysis reveals cellular alterations in psychosis and markers of treatment-resistant schizophrenia. Elife. 2021;10:e58430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Aberg KA, McClay JL, Nerella S, Clark S, Kumar G, Chen W, et al. Methylome-wide association study of schizophrenia: identifying blood biomarker signatures of environmental insults. JAMA Psychiatry. 2014;71:255–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Wang W, Li W, Wu Y, Tian X, Duan H, Li S, et al. Genome-wide DNA methylation and gene expression analyses in monozygotic twins identify potential biomarkers of depression. Transl Psychiatry. 2021;11:416. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Grayson DR, Jia X, Chen Y, Sharma RP, Mitchell CP, Guidotti A, et al. Reelin promoter hypermethylation in schizophrenia. Proc Natl Acad Sci USA. 2005;102:9341–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Abdolmaleky HM, Cheng KH, Russo A, Smith CL, Faraone SV, Wilcox M, et al. Hypermethylation of the reelin (RELN) promoter in the brain of schizophrenic patients: a preliminary report. Am J Med Genet B Neuropsychiatr Genet. 2005;134b:60–66. [DOI] [PubMed] [Google Scholar]
- 53.Nabil Fikri RM, Norlelawati AT, Nour El-Huda AR, Hanisah MN, Kartini A, Norsidah K, et al. Reelin (RELN) DNA methylation in the peripheral blood of schizophrenia. J Psychiatr Res. 2017;88:28–37. [DOI] [PubMed] [Google Scholar]
- 54.Zhou J, Zhou D, Yan T, Chen W, Xie H, Xiong Y. Association between CpG island DNA methylation in the promoter region of RELN and positive and negative types of schizophrenia. J Int Med Res. 2022;50:3000605221100345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Tochigi M, Iwamoto K, Bundo M, Komori A, Sasaki T, Kato N, et al. Methylation status of the reelin promoter region in the brain of schizophrenic patients. Biol Psychiatry. 2008;63:530–3. [DOI] [PubMed] [Google Scholar]
- 56.Bonsch D, Wunschel M, Lenz B, Janssen G, Weisbrod M, Sauer H. Methylation matters? Decreased methylation status of genomic DNA in the blood of schizophrenic twins. Psychiatry Res. 2012;198:533–7. [DOI] [PubMed] [Google Scholar]
- 57.Dong E, Gavin DP, Chen Y, Davis J. Upregulation of TET1 and downregulation of APOBEC3A and APOBEC3C in the parietal cortex of psychotic patients. Transl Psychiatry. 2012;2:e159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Huang HS, Akbarian S. GAD1 mRNA expression and DNA methylation in prefrontal cortex of subjects with schizophrenia. PLoS One. 2007;2:e809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Abdolmaleky HM, Cheng KH, Faraone SV, Wilcox M, Glatt SJ, Gao F, et al. Hypomethylation of MB-COMT promoter is a major risk factor for schizophrenia and bipolar disorder. Hum Mol Genet. 2006;15:3132–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Nohesara S, Ghadirivasfi M, Mostafavi S, Eskandari MR, Ahmadkhaniha H, Thiagalingam S, et al. DNA hypomethylation of MB-COMT promoter in the DNA derived from saliva in schizophrenia and bipolar disorder. J Psychiatr Res. 2011;45:1432–8. [DOI] [PubMed] [Google Scholar]
- 61.Walton E, Liu J, Hass J, White T, Scholz M, Roessner V, et al. MB-COMT promoter DNA methylation is associated with working-memory processing in schizophrenia patients and healthy controls. Epigenetics. 2014;9:1101–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Dai D, Cheng J, Zhou K, Lv Y, Zhuang Q, Zheng R, et al. Significant association between DRD3 gene body methylation and schizophrenia. Psychiatry Res. 2014;220:772–7. [DOI] [PubMed] [Google Scholar]
- 63.Cheng J, Wang Y, Zhou K, Wang L, Li J, Zhuang Q, et al. Male-specific association between dopamine receptor D4 gene methylation and schizophrenia. PLoS One. 2014;9:e89128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Funahashi Y, Yoshino Y, Yamazaki K, Ozaki Y, Mori Y, Mori T, et al. Analysis of methylation and −141C Ins/Del polymorphisms of the dopamine receptor D2 gene in patients with schizophrenia. Psychiatry Res. 2019;278:135–40. [DOI] [PubMed] [Google Scholar]
- 65.Antoniuk S, Bijata M, Ponimaskin E, Wlodarczyk J. Chronic unpredictable mild stress for modeling depression in rodents: meta-analysis of model reliability. Neurosci Biobehav Rev. 2019;99:101–16. [DOI] [PubMed] [Google Scholar]
- 66.Ferland CL, Schrader LA. Regulation of histone acetylation in the hippocampus of chronically stressed rats: a potential role of sirtuins. Neuroscience. 2011;174:104–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.LaPlant Q, Vialou V, Covington HE 3rd, Dumitriu D, Feng J, Warren BL, et al. Dnmt3a regulates emotional behavior and spine plasticity in the nucleus accumbens. Nat Neurosci. 2010;13:1137–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Stankiewicz AM, Swiergiel AH, Lisowski P. Epigenetics of stress adaptations in the brain. Brain Res Bull. 2013;98:76–92. [DOI] [PubMed] [Google Scholar]
- 69.Li M, D’Arcy C, Li X, Zhang T, Joober R, Meng X. What do DNA methylation studies tell us about depression? A systematic review. Transl Psychiatry. 2019;9:68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Uchida S, Hara K, Kobayashi A, Otsuki K, Yamagata H, Hobara T, et al. Epigenetic status of Gdnf in the ventral striatum determines susceptibility and adaptation to daily stressful events. Neuron. 2011;69:359–72. [DOI] [PubMed] [Google Scholar]
- 71.Park C, Rosenblat JD, Brietzke E, Pan Z, Lee Y, Cao B, et al. Stress, epigenetics and depression: a systematic review. Neurosci Biobehav Rev. 2019;102:139–52. [DOI] [PubMed] [Google Scholar]
- 72.Sales AJ, Maciel IS, Suavinha A, Joca SRL. Modulation of DNA methylation and gene expression in rodent cortical neuroplasticity pathways exerts rapid antidepressant-like effects. Mol Neurobiol. 2021;58:777–94. [DOI] [PubMed] [Google Scholar]
- 73.Bang M, Kang JI, Kim SJ, Park JY, Kim KR, Lee SY, et al. Reduced DNA methylation of the oxytocin receptor gene is associated with anhedonia-asociality in women with recent-onset schizophrenia and ultra-high risk for psychosis. Schizophr Bull. 2019;45:1279–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Luo C, Pi X, Hu N, Wang X, Xiao Y, Li S, et al. Subtypes of schizophrenia identified by multi-omic measures associated with dysregulated immune function. Mol Psychiatry. 2021;26:6926–36. [DOI] [PubMed] [Google Scholar]
- 75.Yamagata H, Ogihara H, Matsuo K, Uchida S, Kobayashi A, Seki T, et al. Distinct epigenetic signatures between adult-onset and late-onset depression. Sci Rep. 2021;11:2296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Li Z, He Y, Ma X, Chen X. Epigenetic age analysis of brain in major depressive disorder. Psychiatry Res. 2018;269:621–4. [DOI] [PubMed] [Google Scholar]
- 77.Hodes GE, Pfau ML, Purushothaman I, Ahn HF, Golden SA, Christoffel DJ, et al. Sex differences in nucleus accumbens transcriptome profiles associated with susceptibility versus resilience to subchronic variable stress. J Neurosci. 2015;35:16362–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Watkins JC, Evans RH. Excitatory amino acid transmitters. Annu Rev Pharm Toxicol. 1981;21:165–204. [DOI] [PubMed] [Google Scholar]
- 79.Johnson JW, Ascher P. Glycine potentiates the NMDA response in cultured mouse brain neurons. Nature. 1987;325:529–31. [DOI] [PubMed] [Google Scholar]
- 80.Clements JD, Westbrook GL. Activation kinetics reveal the number of glutamate and glycine binding sites on the N-methyl-D-aspartate receptor. Neuron. 1991;7:605–13. [DOI] [PubMed] [Google Scholar]
- 81.Moriyoshi K, Masu M, Ishii T, Shigemoto R, Mizuno N, Nakanishi S. Molecular cloning and characterization of the rat NMDA receptor. Nature. 1991;354:31–37. [DOI] [PubMed] [Google Scholar]
- 82.Monyer H, Sprengel R, Schoepfer R, Herb A, Higuchi M, Lomeli H, et al. Heteromeric NMDA receptors: molecular and functional distinction of subtypes. Science. 1992;256:1217–21. [DOI] [PubMed] [Google Scholar]
- 83.Das S, Sasaki YF, Rothe T, Premkumar LS, Takasu M, Crandall JE, et al. Increased NMDA current and spine density in mice lacking the NMDA receptor subunit NR3A. Nature. 1998;393:377–81. [DOI] [PubMed] [Google Scholar]
- 84.Sun L, Margolis FL, Shipley MT, Lidow MS. Identification of a long variant of mRNA encoding the NR3 subunit of the NMDA receptor: its regional distribution and developmental expression in the rat brain. FEBS Lett. 1998;441:392–6. [DOI] [PubMed] [Google Scholar]
- 85.Sugihara H, Moriyoshi K, Ishii T, Masu M, Nakanishi S. Structures and properties of seven isoforms of the NMDA receptor generated by alternative splicing. Biochem Biophys Res Commun. 1992;185:826–32. [DOI] [PubMed] [Google Scholar]
- 86.Hollmann M, Boulter J, Maron C, Beasley L, Sullivan J, Pecht G, et al. Zinc potentiates agonist-induced currents at certain splice variants of the NMDA receptor. Neuron. 1993;10:943–54. [DOI] [PubMed] [Google Scholar]
- 87.Mandich P, Schito AM, Bellone E, Antonacci R, Finelli P, Rocchi M, et al. Mapping of the human NMDAR2B receptor subunit gene (GRIN2B) to chromosome 12p12. Genomics. 1994;22:216–8. [DOI] [PubMed] [Google Scholar]
- 88.Kalsi G, Whiting P, Bourdelles BL, Callen D, Barnard EA, Gurling H. Localization of the human NMDAR2D receptor subunit gene (GRIN2D) to 19q13.1-qter, the NMDAR2A subunit gene to 16p13.2 (GRIN2A), and the NMDAR2C subunit gene (GRIN2C) to 17q24-q25 using somatic cell hybrid and radiation hybrid mapping panels. Genomics. 1998;47:423–5. [DOI] [PubMed] [Google Scholar]
- 89.Andersson O, Stenqvist A, Attersand A, von Euler G. Nucleotide sequence, genomic organization, and chromosomal localization of genes encoding the human NMDA receptor subunits NR3A and NR3B. Genomics. 2001;78:178–84. [DOI] [PubMed] [Google Scholar]
- 90.Tovar KR, Westbrook GL. Mobile NMDA receptors at hippocampal synapses. Neuron. 2002;34:255–64. [DOI] [PubMed] [Google Scholar]
- 91.Delint-Ramirez I, Salcedo-Tello P, Bermudez-Rattoni F. Spatial memory formation induces recruitment of NMDA receptor and PSD-95 to synaptic lipid rafts. J Neurochem. 2008;106:1658–68. [DOI] [PubMed] [Google Scholar]
- 92.Quinlan EM, Philpot BD, Huganir RL, Bear MF. Rapid, experience-dependent expression of synaptic NMDA receptors in visual cortex in vivo. Nat Neurosci. 1999;2:352–7. [DOI] [PubMed] [Google Scholar]
- 93.Erreger K, Dravid SM, Banke TG, Wyllie DJ, Traynelis SF. Subunit-specific gating controls rat NR1/NR2A and NR1/NR2B NMDA channel kinetics and synaptic signalling profiles. J Physiol. 2005;563:345–58. Pt 2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Tovar KR, Westbrook GL. The incorporation of NMDA receptors with a distinct subunit composition at nascent hippocampal synapses in vitro. J Neurosci. 1999;19:4180–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Xue M, Zhou SB, Liu RH, Chen QY, Zhuo M, Li XH. NMDA receptor-dependent synaptic depression in potentiated synapses of the anterior cingulate cortex of adult mice. Mol Pain. 2021;17:17448069211018045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Bennett MR, Lagopoulos J. Stress and trauma: BDNF control of dendritic-spine formation and regression. Prog Neurobiol. 2014;112:80–99. [DOI] [PubMed] [Google Scholar]
- 97.Caldeira MV, Melo CV, Pereira DB, Carvalho RF, Carvalho AL, Duarte CB. BDNF regulates the expression and traffic of NMDA receptors in cultured hippocampal neurons. Mol Cell Neurosci. 2007;35:208–19. [DOI] [PubMed] [Google Scholar]
- 98.Minichiello L TrkB signalling pathways in LTP and learning. Nat Rev Neurosci. 2009;10:850–60. [DOI] [PubMed] [Google Scholar]
- 99.Monaco SA, Gulchina Y, Gao WJ. NR2B subunit in the prefrontal cortex: a double-edged sword for working memory function and psychiatric disorders. Neurosci Biobehav Rev. 2015;56:127–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Gao WJ, Yang SS, Mack NR, Chamberlin LA. Aberrant maturation and connectivity of prefrontal cortex in schizophrenia-contribution of NMDA receptor development and hypofunction. Mol Psychiatry. 2022;27:731–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Perszyk RE, DiRaddo JO, Strong KL, Low CM, Ogden KK, Khatri A, et al. GluN2D-containing N-methyl-d-aspartate receptors mediate synaptic transmission in hippocampal interneurons and regulate interneuron activity. Mol Pharmacol. 2016;90:689–702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Monyer H, Burnashev N, Laurie DJ, Sakmann B, Seeburg PH. Developmental and regional expression in the rat brain and functional properties of four NMDA receptors. Neuron. 1994;12:529–40. [DOI] [PubMed] [Google Scholar]
- 103.Wenzel A, Scheurer L, Künzi R, Fritschy JM, Mohler H, Benke D. Distribution of NMDA receptor subunit proteins NR2A, 2B, 2C and 2D in rat brain. Neuroreport. 1995;7:45–48. [PubMed] [Google Scholar]
- 104.Standaert DG, Landwehrmeyer GB, Kerner JA, Penney JB Jr., Young AB. Expression of NMDAR2D glutamate receptor subunit mRNA in neurochemically identified interneurons in the rat neostriatum, neocortex and hippocampus. Brain Res Mol Brain Res. 1996;42:89–102. [DOI] [PubMed] [Google Scholar]
- 105.Rauner C, Kohr G. Triheteromeric NR1/NR2A/NR2B receptors constitute the major N-methyl-D-aspartate receptor population in adult hippocampal synapses. J Biol Chem. 2011;286:7558–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Luo J, Wang Y, Yasuda RP, Dunah AW, Wolfe BB. The majority of N-methyl-D-aspartate receptor complexes in adult rat cerebral cortex contain at least three different subunits (NR1/NR2A/NR2B). Mol Pharmacol. 1997;51:79–86. [DOI] [PubMed] [Google Scholar]
- 107.Gray JA, Shi Y, Usui H, During MJ, Sakimura K, Nicoll RA. Distinct modes of AMPA receptor suppression at developing synapses by GluN2A and GluN2B: single-cell NMDA receptor subunit deletion in vivo. Neuron. 2011;71:1085–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Williams K, Russell SL, Shen YM, Molinoff PB. Developmental switch in the expression of NMDA receptors occurs in vivo and in vitro. Neuron. 1993;10:267–78. [DOI] [PubMed] [Google Scholar]
- 109.Ogden KK, Traynelis SF. New advances in NMDA receptor pharmacology. Trends Pharm Sci. 2011;32:726–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Vyklicky V, Korinek M, Smejkalova T, Balik A, Krausova B, Kaniakova M, et al. Structure, function, and pharmacology of NMDA receptor channels. Physiol Res. 2014;63:S191–203. [DOI] [PubMed] [Google Scholar]
- 111.Krystal JH, Karper LP, Seibyl JP, Freeman GK, Delaney R, Bremner JD, et al. Subanesthetic effects of the noncompetitive NMDA antagonist, ketamine, in humans. Psychotomimetic, perceptual, cognitive, and neuroendocrine responses. Arch Gen Psychiatry. 1994;51:199–214 [DOI] [PubMed] [Google Scholar]
- 112.Malhotra AK, Pinals DA, Weingartner H, Sirocco K, Missar CD, Pickar D, et al. NMDA receptor function and human cognition: the effects of ketamine in healthy volunteers. Neuropsychopharmacology. 1996;14:301–7. [DOI] [PubMed] [Google Scholar]
- 113.Newcomer JW, Farber NB, Jevtovic-Todorovic V, Selke G, Melson AK, Hershey T, et al. Ketamine-induced NMDA receptor hypofunction as a model of memory impairment and psychosis. Neuropsychopharmacology. 1999;20:106–18. [DOI] [PubMed] [Google Scholar]
- 114.Olney JW, Farber NB. NMDA antagonists as neurotherapeutic drugs, psychotogens, neurotoxins, and research tools for studying schizophrenia. Neuropsychopharmacology. 1995;13:335–45. [DOI] [PubMed] [Google Scholar]
- 115.Malhotra AK, Adler CM, Kennison SD, Elman I, Pickar D, Breier A. Clozapine blunts N-methyl-D-aspartate antagonist-induced psychosis: a study with ketamine. Biol psychiatry. 1997;42:664–8. [DOI] [PubMed] [Google Scholar]
- 116.Lahti AC, Weiler MA, Tamara Michaelidis BA, Parwani A, Tamminga CA. Effects of ketamine in normal and schizophrenic volunteers. Neuropsychopharmacology. 2001;25:455–67. [DOI] [PubMed] [Google Scholar]
- 117.Snyder MA, Gao WJ. NMDA receptor hypofunction for schizophrenia revisited: Perspectives from epigenetic mechanisms. Schizophr Res. 2020;217:60–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Adell A. Brain NMDA receptors in schizophrenia and depression. Biomolecules. 2020;10:947. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Coyle JT. NMDA receptor and schizophrenia: a brief history. Schizophr Bull. 2012;38:920–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Catts VS, Lai YL, Weickert CS, Weickert TW, Catts SV. A quantitative review of the postmortem evidence for decreased cortical N-methyl-d-aspartate receptor expression levels in schizophrenia: How can we link molecular abnormalities to mismatch negativity deficits? Biol Psychol. 2016;116:57–67. [DOI] [PubMed] [Google Scholar]
- 121.Singh T, Poterba T, Curtis D, Akil H, Al Eissa M, Barchas JD, et al. Rare coding variants in ten genes confer substantial risk for schizophrenia. Nature. 2022;604:509–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Trubetskoy V, Pardiñas AF, Qi T, Panagiotaropoulou G, Awasthi S, Bigdeli TB, et al. Mapping genomic loci implicates genes and synaptic biology in schizophrenia. Nature. 2022;604:502–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Banerjee A, Wang HY, Borgmann-Winter KE, MacDonald ML, Kaprielian H, Stucky A, et al. Src kinase as a mediator of convergent molecular abnormalities leading to NMDAR hypoactivity in schizophrenia. Mol Psychiatry. 2015;20:1091–1100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.McNally JM, McCarley RW. Gamma band oscillations: a key to understanding schizophrenia symptoms and neural circuit abnormalities. Curr Opin Psychiatry. 2016;29:202–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Antonoudiou P, Tan YL, Kontou G, Upton AL, Mann EO. Parvalbumin and somatostatin interneurons contribute to the generation of hippocampal gamma oscillations. J Neurosci. 2020;40:7668–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Gonzalez-Burgos G, Lewis DA. NMDA receptor hypofunction, parvalbumin-positive neurons, and cortical gamma oscillations in schizophrenia. Schizophr Bull. 2012;38:950–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Pinault D. N-methyl d-aspartate receptor antagonists ketamine and MK-801 induce wake-related aberrant gamma oscillations in the rat neocortex. Biol Psychiatry. 2008;63:730–5. [DOI] [PubMed] [Google Scholar]
- 128.Hong LE, Summerfelt A, Buchanan RW, O’Donnell P, Thaker GK, Weiler MA, et al. Gamma and delta neural oscillations and association with clinical symptoms under subanesthetic ketamine. Neuropsychopharmacology. 2010;35:632–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Aguilar DD, Radzik LK, Schiffino FL, Folorunso OO, Zielinski MR, Coyle JT, et al. Altered neural oscillations and behavior in a genetic mouse model of NMDA receptor hypofunction. Sci Rep. 2021;11:9031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Jami SA, Cameron S, Wong JM, Daly ER, McAllister AK, Gray JA. Increased excitation-inhibition balance and loss of GABAergic synapses in the serine racemase knockout model of NMDA receptor hypofunction. J Neurophysiol. 2021;126:11–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Alvarez RJ, Pafundo DE, Zold CL, Belforte JE. Interneuron NMDA receptor ablation induces hippocampus-prefrontal cortex functional hypoconnectivity after adolescence in a mouse model of schizophrenia. J Neurosci. 2020;40:3304–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Tan Y, Fujita Y, Pu Y, Chang L, Qu Y, Wang X, et al. Repeated intermittent administration of (R)-ketamine during juvenile and adolescent stages prevents schizophrenia-relevant phenotypes in adult offspring after maternal immune activation: a role of TrkB signaling. Eur Arch Psychiatry Clin Neurosci. 2022;272:693–701. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Kegeles LS, Abi-Dargham A, Zea-Ponce Y, Rodenhiser-Hill J, Mann JJ, Van Heertum RL, et al. Modulation of amphetamine-induced striatal dopamine release by ketamine in humans: implications for schizophrenia. Biol Psychiatry. 2000;48:627–40. [DOI] [PubMed] [Google Scholar]
- 134.Tanqueiro SR, Mouro FM, Ferreira CB, Freitas CF, Fonseca-Gomes J, Simoes do Couto F, et al. Sustained NMDA receptor hypofunction impairs brain-derived neurotropic factor signalling in the PFC, but not in the hippocampus, and disturbs PFC-dependent cognition in mice. J Psychopharmacol. 2021;35:730–43. [DOI] [PubMed] [Google Scholar]
- 135.Lopez-Gil X, Artigas F, Adell A. Role of different monoamine receptors controlling MK-801-induced release of serotonin and glutamate in the medial prefrontal cortex: relevance for antipsychotic action. Int J Neuropsychopharmacol. 2009;12:487–99. [DOI] [PubMed] [Google Scholar]
- 136.Crane GE. Cyloserine as an antidepressant agent. Am J Psychiatry. 1959;115:1025–6. [DOI] [PubMed] [Google Scholar]
- 137.Trullas R, Skolnick P. Functional antagonists at the NMDA receptor complex exhibit antidepressant actions. Eur J Pharm. 1990;185:1–10. [DOI] [PubMed] [Google Scholar]
- 138.Berman RM, Cappiello A, Anand A, Oren DA, Heninger GR, Charney DS, et al. Antidepressant effects of ketamine in depressed patients. Biol Psychiatry. 2000;47:351–4. [DOI] [PubMed] [Google Scholar]
- 139.Hu YD, Xiang YT, Fang JX, Zu S, Sha S, Shi H, et al. Single i.v. ketamine augmentation of newly initiated escitalopram for major depression: results from a randomized, placebo-controlled 4-week study. Psychol Med. 2016;46:623–35. [DOI] [PubMed] [Google Scholar]
- 140.Daly EJ, Trivedi MH, Janik A, Li H, Zhang Y, Li X, et al. Efficacy of esketamine nasal spray plus oral antidepressant treatment for relapse prevention in patients with treatment-resistant depression: a randomized clinical trial. JAMA Psychiatry. 2019;76:893–903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Popova V, Daly EJ, Trivedi M, Cooper K, Lane R, Lim P, et al. Efficacy and safety of flexibly dosed esketamine nasal spray combined with a newly initiated oral antidepressant in treatment-resistant depression: a randomized double-blind active-controlled study. Am J Psychiatry. 2019;176:428–38. [DOI] [PubMed] [Google Scholar]
- 142.Vázquez GH, Bahji A, Undurraga J, Tondo L, Baldessarini RJ. Efficacy and tolerability of combination treatments for major depression: antidepressants plus second-generation antipsychotics vs. esketamine vs. lithium. J Psychopharmacol. 2021;35:890–900. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Jelen LA, Young AH, Stone JM. Ketamine: a tale of two enantiomers. J Psychopharmacol. 2021;35:109–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Breier A, Malhotra AK, Pinals DA, Weisenfeld NI, Pickar D. Association of ketamine-induced psychosis with focal activation of the prefrontal cortex in healthy volunteers. Am J Psychiatry. 1997;154:805–11. [DOI] [PubMed] [Google Scholar]
- 145.Moghaddam B, Adams B, Verma A, Daly D. Activation of glutamatergic neurotransmission by ketamine: a novel step in the pathway from NMDA receptor blockade to dopaminergic and cognitive disruptions associated with the prefrontal cortex. J Neurosci. 1997;17:2921–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Ebert B, Mikkelsen S, Thorkildsen C, Borgbjerg FM. Norketamine, the main metabolite of ketamine, is a non-competitive NMDA receptor antagonist in the rat cortex and spinal cord. Eur J Pharm. 1997;333:99–104. [DOI] [PubMed] [Google Scholar]
- 147.Yang C, Shirayama Y, Zhang JC, Ren Q, Yao W, Ma M, et al. R-ketamine: a rapid-onset and sustained antidepressant without psychotomimetic side effects. Transl Psychiatry. 2015;5:e632. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Fukumoto K, Toki H, Iijima M, Hashihayata T, Yamaguchi JI, Hashimoto K, et al. Antidepressant potential of (R)-ketamine in rodent models: comparison with (S)-ketamine. J Pharm Exp Ther. 2017;361:9–16. [DOI] [PubMed] [Google Scholar]
- 149.Chang L, Zhang K, Pu Y, Qu Y, Wang SM, Xiong Z, et al. Comparison of antidepressant and side effects in mice after intranasal administration of (R,S)-ketamine, (R)-ketamine, and (S)-ketamine. Pharm Biochem Behav. 2019;181:53–59. [DOI] [PubMed] [Google Scholar]
- 150.Bonaventura J, Lam S, Carlton M, Boehm MA, Gomez JL, Solis O, et al. Pharmacological and behavioral divergence of ketamine enantiomers: implications for abuse liability. Mol Psychiatry. 2021;26:6704–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Hashimoto K, Kakiuchi T, Ohba H, Nishiyama S, Tsukada H. Reduction of dopamine D2/3 receptor binding in the striatum after a single administration of esketamine, but not R-ketamine: a PET study in conscious monkeys. Eur Arch Psychiatry Clin Neurosci. 2017;267:173–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Masaki Y, Kashiwagi Y, Watabe H, Abe K. (R)- and (S)-ketamine induce differential fMRI responses in conscious rats. Synapse. 2019;73:e22126. [DOI] [PubMed] [Google Scholar]
- 153.Miller OH, Yang L, Wang CC, Hargroder EA, Zhang Y, Delpire E, et al. GluN2B-containing NMDA receptors regulate depression-like behavior and are critical for the rapid antidepressant actions of ketamine. Elife. 2014;3:e03581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Jang S, Suh SH, Yoo HS, Lee YM, Oh S. Changes in iNOS, GFAP and NR1 expression in various brain regions and elevation of sphingosine-1-phosphate in serum after immobilized stress. Neurochem Res. 2008;33:842–51. [DOI] [PubMed] [Google Scholar]
- 155.Bartanusz V, Aubry JM, Pagliusi S, Jezova D, Baffi J, Kiss JZ. Stress-induced changes in messenger RNA levels of N-methyl-D-aspartate and AMPA receptor subunits in selected regions of the rat hippocampus and hypothalamus. Neuroscience. 1995;66:247–52. [DOI] [PubMed] [Google Scholar]
- 156.Fitzgerald LW, Ortiz J, Hamedani AG, Nestler EJ. Drugs of abuse and stress increase the expression of GluR1 and NMDAR1 glutamate receptor subunits in the rat ventral tegmental area: common adaptations among cross-sensitizing agents. J Neurosci. 1996;16:274–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Masrour FF, Peeri M, Azarbayjani MA, Hosseini MJ. Voluntary exercise during adolescence mitigated negative the effects of maternal separation stress on the depressive-like behaviors of adult male rats: role of NMDA receptors. Neurochem Res. 2018;43:1067–74. [DOI] [PubMed] [Google Scholar]
- 158.Dong BE, Chen H, Sakata K. BDNF deficiency and enriched environment treatment affect neurotransmitter gene expression differently across ages. J Neurochem. 2020;154:41–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Sathyanesan M, Haiar JM, Watt MJ, Newton SS. Restraint stress differentially regulates inflammation and glutamate receptor gene expression in the hippocampus of C57BL/6 and BALB/c mice. Stress. 2017;20:197–204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Weiland NG, Orchinik M, Tanapat P. Chronic corticosterone treatment induces parallel changes in N-methyl-D-aspartate receptor subunit messenger RNA levels and antagonist binding sites in the hippocampus. Neuroscience. 1997;78:653–62. [DOI] [PubMed] [Google Scholar]
- 161.Pacheco A, Aguayo FI, Aliaga E, Munoz M, Garcia-Rojo G, Olave FA, et al. Chronic stress triggers expression of immediate early genes and differentially affects the expression of AMPA and NMDA subunits in dorsal and ventral hippocampus of rats. Front Mol Neurosci. 2017;10:244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Tordera RM, Garcia-García AL, Elizalde N, Segura V, Aso E, Venzala E, et al. Chronic stress and impaired glutamate function elicit a depressive-like phenotype and common changes in gene expression in the mouse frontal cortex. Eur Neuropsychopharmacol. 2011;21:23–32. [DOI] [PubMed] [Google Scholar]
- 163.Lee YA, Goto Y. Chronic stress modulation of prefrontal cortical NMDA receptor expression disrupts limbic structure-prefrontal cortex interaction. Eur J Neurosci. 2011;34:426–36. [DOI] [PubMed] [Google Scholar]
- 164.Fachim HA, Loureiro CM, Corsi-Zuelli F, Shuhama R, Louzada-Junior P, Menezes PR, et al. GRIN2B promoter methylation deficits in early-onset schizophrenia and its association with cognitive function. Epigenomics. 2019;11:401–10. [DOI] [PubMed] [Google Scholar]
- 165.Loureiro CM, Fachim HA, Corsi-Zuelli F, Shuhama R, Menezes PR, Dalton CF, et al. The relationship of childhood trauma and DNA methylation of NMDA receptor genes in first-episode schizophrenia. Epigenomics. 2021;13:927–37. [DOI] [PubMed] [Google Scholar]
- 166.Loureiro CM, Fachim HA, Corsi-Zuelli F, Shuhama R, Joca S, Menezes PR, et al. Epigenetic-mediated N-methyl-D-aspartate receptor changes in the brain of isolated reared rats. Epigenomics. 2020;12:1983–97. [DOI] [PubMed] [Google Scholar]
- 167.Loureiro CM, Fachim HA, Harte MK, Dalton CF, Reynolds GP, Subchronic PCP. effects on DNA methylation and protein expression of NMDA receptor subunit genes in the prefrontal cortex and hippocampus of female rats. J Psychopharmacol. 2022;36:238–44. [DOI] [PubMed] [Google Scholar]
- 168.Gulchina Y, Xu SJ, Snyder MA, Elefant F, Gao WJ. Epigenetic mechanisms underlying NMDA receptor hypofunction in the prefrontal cortex of juvenile animals in the MAM model for schizophrenia. J Neurochemistry. 2017;143:320–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Latusz J, Maćkowiak M. Early-life blockade of NMDA receptors induces epigenetic abnormalities in the adult medial prefrontal cortex: possible involvement in memory impairment in trace fear conditioning. Psychopharmacology. 2020;237:231–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Bharadwaj R, Peter Cyril J, Jiang Y, Roussos P, Vogel-Ciernia A, Shen EY, et al. Conserved higher-order chromatin regulates NMDA receptor gene expression and cognition. Neuron. 2014;84:997–1008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Jiang Y, Jakovcevski M, Bharadwaj R, Connor C, Schroeder FA, Lin CL, et al. Setdb1 histone methyltransferase regulates mood-related behaviors and expression of the NMDA receptor subunit NR2B. J Neurosci. 2010;30:7152–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Weder N, Zhang H, Jensen K, Yang BZ, Simen A, Jackowski A, et al. Child abuse, depression, and methylation in genes involved with stress, neural plasticity, and brain circuitry. J Am Acad Child Adolesc Psychiatry. 2014;53:417–424 e415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Montalvo-Ortiz JL, Bordner KA, Carlyle BC, Gelernter J, Simen AA, Kaufman J. The role of genes involved in stress, neural plasticity, and brain circuitry in depressive phenotypes: Convergent findings in a mouse model of neglect. Behav Brain Res. 2016;315:71–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Kaut O, Schmitt I, Hofmann A, Hoffmann P, Schlaepfer TE, Wullner U, et al. Aberrant NMDA receptor DNA methylation detected by epigenome-wide analysis of hippocampus and prefrontal cortex in major depression. Eur Arch Psychiatry Clin Neurosci. 2015;265:331–41. [DOI] [PubMed] [Google Scholar]
- 175.Reiff CM, Richman EE, Nemeroff CB, Carpenter LL, Widge AS, Rodriguez CI, et al. Psychedelics and psychedelic-assisted psychotherapy. Am J Psychiatry. 2020;177:391–410. [DOI] [PubMed] [Google Scholar]
- 176.Inserra A, De Gregorio D, Gobbi G. Psychedelics in psychiatry: neuroplastic, immunomodulatory, and neurotransmitter mechanisms. Pharmacol Rev. 2021;73:202–77. [DOI] [PubMed] [Google Scholar]
- 177.Shao LX, Liao C, Gregg I, Davoudian PA, Savalia NK, Delagarza K, et al. Psilocybin induces rapid and persistent growth of dendritic spines in frontal cortex in vivo. Neuron. 2021;109:2535–2544.e2534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Jefferson SJ, Gregg I, Dibbs M, Liao C, Wu H, Davoudian PA, et al. 5-MeO-DMT modifies innate behaviors and promotes structural neural plasticity in mice. Neuropsychopharmacology. 2023;48:1257–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Ly C, Greb AC, Cameron LP, Wong JM, Barragan EV, Wilson PC, et al. Psychedelics promote structural and functional neural plasticity. Cell Rep. 2018;23:3170–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Doss MK, Považan M, Rosenberg MD, Sepeda ND, Davis AK, Finan PH, et al. Psilocybin therapy increases cognitive and neural flexibility in patients with major depressive disorder. Transl Psychiatry. 2021;11:574. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Skosnik PD, Sloshower J, Safi-Aghdam H, Pathania S, Syed S, Pittman B, et al. Sub-acute effects of psilocybin on EEG correlates of neural plasticity in major depression: relationship to symptoms. J Psychopharmacol. 2023;37:687–97. [DOI] [PubMed] [Google Scholar]
- 182.Moliner R, Girych M, Brunello CA, Kovaleva V, Biojone C, Enkavi G, et al. Psychedelics promote plasticity by directly binding to BDNF receptor TrkB. Nat Neurosci. 2023;26:1032–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Hesselgrave N, Troppoli TA, Wulff AB, Cole AB, Thompson SM. Harnessing psilocybin: antidepressant-like behavioral and synaptic actions of psilocybin are independent of 5-HT2R activation in mice. Proc Natl Acad Sci USA. 2021;118:e2022489118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Nardou R, Sawyer E, Song YJ, Wilkinson M, Padovan-Hernandez Y, de Deus JL, et al. Psychedelics reopen the social reward learning critical period. Nature. 2023;618:790–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Nardou R, Lewis EM, Rothhaas R, Xu R, Yang A, Boyden E, et al. Oxytocin-dependent reopening of a social reward learning critical period with MDMA. Nature. 2019;569:116–20. [DOI] [PubMed] [Google Scholar]
- 186.Gerra MC, Jayanthi S, Manfredini M, Walther D, Schroeder J, Phillips KA, et al. Gene variants and educational attainment in cannabis use: mediating role of DNA methylation. Transl Psychiatry. 2018;8:23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Clark SL, Chan R, Zhao M, Xie LY, Copeland WE, Aberg KA, et al. Methylomic investigation of problematic adolescent cannabis use and its negative mental health consequences. J Am Acad Child Adolesc Psychiatry. 2021;60:1524–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Markunas CA, Hancock DB, Xu Z, Quach BC, Fang F, Sandler DP, et al. Epigenome-wide analysis uncovers a blood-based DNA methylation biomarker of lifetime cannabis use. Am J Med Genet B Neuropsychiatr Genet. 2021;186:173–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Murphy SK, Itchon-Ramos N, Visco Z, Huang Z, Grenier C, Schrott R, et al. Cannabinoid exposure and altered DNA methylation in rat and human sperm. Epigenetics. 2018;13:1208–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Schrott R, Rajavel M, Acharya K, Huang Z, Acharya C, Hawkey A, et al. Sperm DNA methylation altered by THC and nicotine: vulnerability of neurodevelopmental genes with bivalent chromatin. Sci Rep. 2020;10:16022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Wanner NM, Colwell M, Drown C, Faulk C. Subacute cannabidiol alters genome-wide DNA methylation in adult mouse hippocampus. Environ Mol Mutagen. 2020;61:890–900. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Wanner NM, Colwell M, Drown C, Faulk C. Developmental cannabidiol exposure increases anxiety and modifies genome-wide brain DNA methylation in adult female mice. Clin Epigenetics. 2021;13:4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Watson CT, Szutorisz H, Garg P, Martin Q, Landry JA, Sharp AJ, et al. Genome-wide DNA methylation profiling reveals epigenetic changes in the rat nucleus accumbens associated with cross-generational effects of adolescent THC exposure. Neuropsychopharmacology. 2015;40:2993–3005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Cheng MC, Hsu SH, Chen CH. Chronic methamphetamine treatment reduces the expression of synaptic plasticity genes and changes their DNA methylation status in the mouse brain. Brain Res. 2015;1629:126–34. [DOI] [PubMed] [Google Scholar]
- 195.Inserra A, Campanale A, Cheishvili D, Dymov S, Wong A, Marcal N, et al. Modulation of DNA methylation and protein expression in the prefrontal cortex by repeated administration of D-lysergic acid diethylamide (LSD): Impact on neurotropic, neurotrophic, and neuroplasticity signaling. Prog Neuropsychopharmacol Biol Psychiatry. 2022;119:110594. [DOI] [PubMed] [Google Scholar]
- 196.Sun L, Verkaik-Schakel RN, Biber K, Plösch T, Serchov T. Antidepressant treatment is associated with epigenetic alterations of Homer1 promoter in a mouse model of chronic depression. J Affect Disord. 2021;279:501–9. [DOI] [PubMed] [Google Scholar]
- 197.Ju LS, Yang JJ, Lei L, Xia JY, Luo D, Ji MH, et al. The combination of long-term ketamine and extinction training contributes to fear erasure by bdnf methylation. Front Cell Neurosci. 2017;11:100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.de la Fuente Revenga M, Zhu B, Guevara CA, Naler LB, Saunders JM, Zhou Z, et al. Prolonged epigenomic and synaptic plasticity alterations following single exposure to a psychedelic in mice. Cell Rep. 2021;37:109836. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Cameron LP, Tombari RJ, Lu J, Pell AJ, Hurley ZQ, Ehinger Y, et al. A non-hallucinogenic psychedelic analogue with therapeutic potential. Nature. 2021;589:474–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Belforte JE, Zsiros V, Sklar ER, Jiang Z, Yu G, Li Y, et al. Postnatal NMDA receptor ablation in corticolimbic interneurons confers schizophrenia-like phenotypes. Nat Neurosci. 2010;13:76–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Nakao K, Jeevakumar V, Jiang SZ, Fujita Y, Diaz NB, Pretell Annan CA, et al. Schizophrenia-like dopamine release abnormalities in a mouse model of NMDA receptor hypofunction. Schizophr Bull. 2019;45:138–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Nakao K, Singh M, Sapkota K, Hagler BC, Hunter RN, Raman C, et al. GSK3beta inhibition restores cortical gamma oscillation and cognitive behavior in a mouse model of NMDA receptor hypofunction relevant to schizophrenia. Neuropsychopharmacology. 2020;45:2207–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Gunasekaran S, Jacob RS, Omkumar RV. Differential expression of miR-148b, miR-129–2 and miR-296 in animal models of schizophrenia-Relevance to NMDA receptor hypofunction. Neuropharmacology. 2022;210:109024. [DOI] [PubMed] [Google Scholar]
- 204.Rodriguez-Munoz M, Sanchez-Blazquez P, Callado LF, Meana JJ, Garzon-Nino J. Schizophrenia and depression, two poles of endocannabinoid system deregulation. Transl Psychiatry. 2017;7:1291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Karolewicz B, Szebeni K, Gilmore T, Maciag D, Stockmeier CA, Ordway GA. Elevated levels of NR2A and PSD-95 in the lateral amygdala in depression. Int J Neuropsychopharmacol. 2009;12:143–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Karolewicz B, Stockmeier CA, Ordway GA. Elevated levels of the NR2C subunit of the NMDA receptor in the locus coeruleus in depression. Neuropsychopharmacology. 2005;30:1557–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Chandley MJ, Szebeni A, Szebeni K, Crawford JD, Stockmeier CA, Turecki G, et al. Elevated gene expression of glutamate receptors in noradrenergic neurons from the locus coeruleus in major depression. Int J Neuropsychopharmacol. 2014;17:1569–78. [DOI] [PubMed] [Google Scholar]
- 208.Gray AL, Hyde TM, Deep-Soboslay A, Kleinman JE, Sodhi MS. Sex differences in glutamate receptor gene expression in major depression and suicide. Mol Psychiatry. 2015;20:1057–68. [DOI] [PubMed] [Google Scholar]
- 209.Feyissa AM, Chandran A, Stockmeier CA, Karolewicz B. Reduced levels of NR2A and NR2B subunits of NMDA receptor and PSD-95 in the prefrontal cortex in major depression. Prog Neuropsychopharmacol Biol psychiatry. 2009;33:70–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Beneyto M, Kristiansen LV, Oni-Orisan A, McCullumsmith RE, Meador-Woodruff JH. Abnormal glutamate receptor expression in the medial temporal lobe in schizophrenia and mood disorders. Neuropsychopharmacology. 2007;32:1888–902. [DOI] [PubMed] [Google Scholar]
- 211.Beneyto M, Meador-Woodruff JH. Lamina-specific abnormalities of NMDA receptor-associated postsynaptic protein transcripts in the prefrontal cortex in schizophrenia and bipolar disorder. Neuropsychopharmacology. 2008;33:2175–86. [DOI] [PubMed] [Google Scholar]
