Download as pdf or txt
Download as pdf or txt
You are on page 1of 18

Molecular Neurobiology

https://doi.org/10.1007/s12035-020-02145-4

Modulation of DNA Methylation and Gene Expression in Rodent


Cortical Neuroplasticity Pathways Exerts Rapid Antidepressant-Like
Effects
Amanda J. Sales 1,2 & Izaque S. Maciel 1 & Angélica C. D. R. Suavinha 3 & Sâmia R. L. Joca 3,4,5

Received: 21 April 2020 / Accepted: 22 September 2020


# Springer Science+Business Media, LLC, part of Springer Nature 2020

Abstract
Background Stress increases DNA methylation, primarily a suppressive epigenetic mechanism catalyzed by DNA methyltrans-
ferases (DNMT), and decreases the expression of genes involved in neuronal plasticity and mood regulation. Despite chronic
antidepressant treatment decreases stress-induced DNA methylation, it is not known whether inhibition of DNMT would convey
rapid antidepressant-like effects.
Aim This work tested such a hypothesis and evaluated whether a behavioral effect induced by DNMT inhibitors (DNMTi)
corresponds with changes in DNA methylation and transcript levels in genes consistently associated with the neurobiology of
depression and synaptic plasticity (BDNF, TrkB, 5-HT1A, NMDA, and AMPA).
Methods Male Wistar rats received intraperitoneal (i.p.) injection of two pharmacologically different DNMTi (5-AzaD 0.2 and
0.6 mg/kg or RG108 0.6 mg/kg) or vehicle (1 ml/kg), 1 h or 7 days before the learned helplessness test (LH). DNA methylation in
target genes and the correspondent transcript levels were measured in the hippocampus (HPC) and prefrontal cortex (PFC) using
meDIP-qPCR. In parallel separate groups, the antidepressant-like effect of 5-AzaD and RG108 was investigated in the forced
swimming test (FST). The involvement of cortical BDNF-TrkB-mTOR pathways was assessed by intra-ventral medial PFC
(vmPFC) injections of rapamycin (mTOR inhibitor), K252a (TrkB receptor antagonist), or vehicle (0.2 μl/side).
Results We found that both 5-AzaD and RG108 acutely and 7 days before the test decreased escape failures in the LH. LH stress
increased DNA methylation and decreased transcript levels of BDNF IV and TrkB in the PFC, effects that were not significantly
attenuated by RG108 treatment. The systemic administration of 5-AzaD (0.2 mg/kg) and RG108 (0.2 mg/kg) induced an
antidepressant-like effect in FST, which was, however, attenuated by TrkB and mTOR inhibition into the vmPFC.

Highlights
• DNMT inhibitors (5-AzaD and RG108) induce antidepressant-like
properties in rats
• Acute DNA methylation inhibition promotes rapid and sustained effects
in rats
• DNMT inhibitors increase BDNF-TrkB-mTOR signaling in the rat pre-
frontal cortex
Electronic supplementary material The online version of this article
(https://doi.org/10.1007/s12035-020-02145-4) contains supplementary
material, which is available to authorized users.

* Amanda J. Sales 3
Department of BioMolecular Sciences, School of Pharmaceutical
amanda.sales@usp.br Sciences of Ribeirão Preto, University of São Paulo, Ribeirão
Preto, SP, Brazil
* Sâmia R. L. Joca
samia@usp.br
4
Translational Neuropsychiatry Unit, Department of Clinical
1
Department of Pharmacology, School of Medicine of Ribeirão Preto, Medicine, Aarhus University, Aarhus, Denmark
University of São Paulo, Ribeirão Preto, SP, Brazil
5
2
FMRP-USP, Av Bandeirantes, 3900, Ribeirão Preto, SP 14049-900, FCFRP-USP, Av Café, sn, Monte Alegre, Ribeirão
Brazil Preto, SP 14040-903, Brazil
Mol Neurobiol

Conclusion These findings suggest that acute inhibition of stress-induced DNA methylation promotes rapid and sustained
antidepressant effects associated with increased BDNF-TrkB-mTOR signaling in the PFC.

Keywords DNA methylation . DNMT . Antidepressant . RG108 . 5-AzaD . RNAm

Introduction Decreased signaling through AMPAR has, thus, been linked


to impaired BDNF signaling and depression neurobiology.
Major depressive disorder (MDD) is a complex psychiatric Equally relevant to regulate BDNF levels and the antide-
disorder with a lifetime prevalence of approximately 20% pressant effect is the serotonergic 5-HT1A receptor [75–77].
[1] in which the stress is a recognized risk factor [2]. MDD Reduced levels of 5-HT1A have been described in stressed
is characterized by several symptoms including anhedonia, animals and depressed humans [78–83]. In addition, increased
depressive mood, and suicidal thoughts [3]. Previous studies 5-HT1A signaling is necessary for antidepressant action [84]
strongly suggest that the neurobiology underlying MDD in- and the activation of cortical and hippocampal 5-HT 1A
volves genetic and epigenetic factors [4]. Candidate genes heteroreceptors results in antidepressant-like effects [85–88].
consistently implicated in the MDD and in the mechanism Several genes associated with depression and chronic anti-
action of antidepressant drugs include those coding for depressant treatment, including the ones mentioned above,
neurotrophins, as well as glutamate and serotonin signaling. have their expression levels modulated by epigenetic mecha-
Amongst neurotrophins, brain-derived neurotrophic factor nisms, such as DNA methylation [89–94]. Catalyzed by DNA
(BDNF) plays a central role in depression neurobiology. methyltransferase enzymes (DNMT), DNA methylation re-
Reduced levels of BDNF and TrkB [5], its main receptor, have fers to the addition of a methyl group to the C5 position of
been described in the prefrontal cortex (PFC) and the hippo- cytosine in cytosine-phosphate-guanine dinucleotides, leading
campus (HPC) of depressed patients [6–10] and stressed ani- to changes in DNA packing and chromatin structure, thereby
mals [11–14]. This is thought to account for the impaired modulating gene expression without altering the original
neuroplasticity described in stress and depression [15, 16]. DNA sequence [95]. DNA methylation is believed to be a
Accordingly, chronic antidepressant treatment increases key mechanism by which environmental experiences can
BDNF and TrkB signaling in the PFC and HPC and restores modify gene expression, resulting in long-lasting changes in
stress-induced impaired neuroplasticity [17, 18]. Moreover, protein availability and brain function [92, 96].
infusion of BDNF or TrkB overexpression into those brain Interestingly, chronic antidepressant treatment has been
regions promotes antidepressant-like effects [11, 12, 19–29]. shown to reverse stress-induced changes in DNA methylation
The blockade of BDNF-TrkB signaling abrogates the behav- of genes believed to be relevant to the neurobiology of depres-
ioral effect of antidepressants, thus indicating that the upreg- sion [97–99]. Moreover, recovery from clinical depression is
ulation of BDNF may be a central mechanism in the antide- associated with changes in DNMT levels in blood cells [100],
pressant effect [25, 30–32]. pointing to a central role for DNA methylation in the antide-
Significant abnormalities in glutamatergic signaling, which pressant response. In accordance with those findings, we and
is crucial in regulating activity-dependent BDNF expression others have shown that direct inhibition of DNA methylation
and neuroplasticity [33, 34], are also described in depressed by DNMT inhibitors (DNMTi), such as 5-AzaC, 5-AzaD, and
patients [35–45]. Increased levels of NMDA receptors, partic- RG108, results in antidepressant-like effects in preclinical
ularly NMDA2a, have been described in the HPC of rodents tests [101–105].
exposed to stress models of depression [46–53]. On the other In this regard, increased DNA methylation in Bdnf promot-
hand, chronic antidepressant treatment attenuates NMDA2a er, along with reduced mRNA and protein levels, has been
levels and its inactivation or pharmacological blockade pro- described in conditions of stress and depression [106–111].
motes antidepressant properties in animals and depressed Keller et al. (2011) demonstrated also that the hypermethyla-
humans [54–57]. In contrast to NMDA receptors, antidepres- tion in the TrkB (tyrosine kinase B) receptor gene, which
sants increase the signaling mediated by AMPA receptor encodes for the major target of mature BDNF, and reduction
(AMPAR) in the HPC and PFC [58–60]. Of note, the rapid in TrkB protein levels is present in the cortex and HPC of
and sustained antidepressant effects induced by ketamine have suicidal individuals [112, 113]. Similarly, methylome-wide
been associated with NMDA blockade and AMPAR activa- association studies have found MDD-methylation associa-
tion [55, 57, 61–63], with subsequent release of BDNF, and tions in pathways linked to neurotrophin signaling [110] and
activation of the mechanistic target of rapamycin (mTOR) in neuronal plasticity [114].
the medial PFC (mPFC) [64–71]. Interestingly, animals ex- Neurochemical alterations described in MDD may be a
posed to chronic stress and MDD patients show reduced result of epigenetic mechanisms since the expression of
AMPA1 expression in the cortex and HPC [60, 72–74]. NMDA, AMPA, and 5-HT1A is also regulated by DNA
Mol Neurobiol

methylation [115–117]. In fact, an important interplay be- Stereotaxic Surgery and Brain Microinjection
tween DNA methylation and these signaling mechanisms is
thought to be crucial in the control of synaptic plasticity [118, The stereotaxic surgery and brain microinjection were per-
119], which is dysfunctional in stressed animals as well as in formed as described earlier [123]. Briefly, rats were anesthe-
depressed patients [60, 77, 120]. tized with 2,2,2 tribromoethanol 2.5% (i.p., 1 ml/kg) and fixed
Interestingly, activation of 5-HT1A, NMDA2a, AMPA, in the stereotaxic equipment. Two stainless steel guide cannu-
and TrkB receptors have been proven to be central mecha- las (23 G1) were implanted bilaterally 1 mm above the
nisms associated with fast antidepressant effect [67, 121, vmPFC (coordinates AP + 3.3 mm from bregma, ML +
122]. Numerous studies have shown that antidepressants at- 1.9 mm from the medial suture, and DV − 2.4 mm from the
tenuate stress-induced DNA methylation and increase the ex- skull with a lateral inclination of 22°) according to the rat brain
pression of BDNF, TrkB, 5-HT1A, and AMPA in the brain atlas [124]. The cannulas were fixed to the skull using jew-
[98, 102, 103]. However, it is not known, whether disinhibi- eler’s screws and acrylic dental cement. After the surgery, rats
tion of DNA methylation could elicit fast antidepressant ef- received an intramuscular injection of oxytetracycline antibi-
fects, and whether this effect may be accompanied by in- otic (200 mg/kg) and subcutaneous injection of flunixin
creased expression of BDNF, TrkB, 5-HT1A, NMDA, and meglumine anti-inflammatory (50 mg/kg). The animals were
AMPA. housed in Plexiglas boxes (2 rats/box) and observed daily
The aim of this study, therefore, was to test the hypothesis during a week (recovery period), after which the behavioral
that the pharmacological inhibition of DNA methylation by procedures were initiated.
DNMT inhibitor drugs would promote fast and sustained an- For microinjection, a stainless steel injection cannula was
tidepressant effects, which could be associated with increased connected via polyethylene tubing (PE-10) to a 2-μl Hamilton
expression of genes associated with BDNF and TrkB signal- syringe. The injection cannula was 1 mm longer than the
ing pathway. guide cannula to reach the mPFC microinjection site. Drugs
were slowly injected into the intended site (0.2 μl each site)
over a minute to allow diffusion.
Materials and Methods At the end of the experiments, the rats were anesthetized
with chloral hydrate 5% (i.p., 1 ml/kg), and 0.5 μl of 1%
Animals Evan’s blue dye was bilaterally injected into the vmPFC as a
site marker. The brains were perfused with 10% formalin and
Male Wistar rats (250–300 g, 7–8 weeks old) were pur- they were postfixed for 24 h at 4 °C. Brain sections of 40 mm
chased from the animal facility of the University of São were cut using a cryostat (CM-1900; Leica, Wetzlar,
Paulo (Campus of Ribeirão Preto) and brought to the Germany). The injection sites were determined using the rat
animal house associated with the Laboratory of brain atlas of Paxinos and Watson (2013) as a reference
Neuropsychopharmacology, where they remained for (Fig. 1).
1 week before the start of the experiments. The animals
were kept under standard conditions of temperature (24 Forced Swimming Test
± 1 °C) and 12-h/12-h light/dark cycle (lights on at
06:00 AM), with free access to food pellets (Nuvilab The forced swimming test (FST) used was conducted as pre-
CR1, Quintia S.A. Brazil) and tap water. The allocation viously described [101]. Rats were individually placed to
was randomly made in pairs in acrylic boxes (41 × 34 × swim in acrylic cylinders (height 40 cm, diameter 30 cm),
16 cm) covered with 3 cm of shavings that were containing 25 cm of water with controlled temperature (24 ±
changed three times per week. During the LH test, the 1 °C), during 15 min (pretest, PT, session). At the end of the
rats were singly housed (between pretest and test ses- session, the animals were gently dried and returned to their
sions). Handling and behavioral testing were performed home cage. Twenty-four hours later, the animals were submit-
between 8 AM and 2 PM. ted to the swimming test (T) session for 5 min in which the
All experimental procedures were in accordance with immobility time was registered. The immobility was defined
the guidelines of the Brazilian College of Animal as the absence of animal movements except those necessary to
Experimentation (COBEA), which are compliant with hold head above the water. The water was changed after each
international laws and policies. All efforts were made trial to avoid the influence of alarm substances [125].
to minimize animal suffering and reduce the number
of animals used. All behavioral protocols were approved Open Field Test
by the local ethical committee for the use of animals
(CEUA n° 12.1.235.53.2 and n° 10.1.136.53.2). A total Rats were individually placed in a square open field arena
of 452 animals were used in this study. (70 × 70 × 40 cm) and allowed to freely explore it for 5 min.
Mol Neurobiol

The exploratory activity was videotaped and the number of


crossings between the quadrants of the arena was measured.
Before each test, the open field was cleaned with 70% ethanol
solution.

Learned Helplessness

The experiments were done in acrylic boxes of active avoid-


ance for rats divided in two compartments (307 × 333 ×
540 mm; EP11 model, Insight, Brazil) separated by a central
wall with an opening communicating the two compartments.
The floor consisted of stainless steel grids through which
footshocks were applied to the animals. In the first day (day
1), rats were exposed to the pretest (PT) session in which they
received inescapable footshocks (IS, stressed group, 40
shocks, 0.4 mA, duration of 10s, in intervals from 30 to
90 s) or no shocks (30 min habituation in the shuttle box,
Hab). Six days later (day 7), all animals were submitted to
the test session (T, 30 escapable footshocks, ES; 0.4 mA, du-
ration of 10 s, preceded by sound stimulus during 5 s, 60 dB,
670 Hz), when the following behaviors were registered: the
number of escapes/avoidance, number of escape failures, and
number of intertrial crossings [103]. During testing, animals
were able to interrupt the shock delivery/escape by crossing to
the other side of the chamber. If the animal failed to escape
during the shock, the trial was registered as an escape failure.

Collection and Sample Processing

Animals were euthanized at indicated times after the experi-


ments by decapitation preceded by anesthesia with 5% chloral
hydrate (10 ml/kg). Dorsal HPC, ventral HPC, and vmPFC
(including prelimbic and infralimbic cortices) were rapidly
dissected on a cooled dish and stored at − 80 °C for further
analysis. Additionally, the naïve group (no stress nor pharma-
cological treatment) was run in parallel as a control for the
molecular analyses. The brain samples were homogenized in
lysis buffer (137 mM NaCl, 20 mM Tris-HCl pH 8.0, 10%
glycerol). One-half of the sample was used for DNA methyl-
ation analysis and the other half for mRNA analysis.

mRNA Expression by Quantitative RT-PCR

For quantitative PCR (qPCR), total mRNA of the sample was


extracted using a kit (SV Total RNA Isolation System,
Fig. 1 Photomicrographs of a representative histological section and Promega) according to the manufacturer’s instructions and
canulae-tip placements within the ventral medial prefrontal cortex treated with DNAse I (provided in the kit). The amount of
(vmPFC). a Representative coronal brain sections of the rat brain show- RNA was quantified using a nanophotometer (P360 Implen)
ing bilateral microinjection sites in the vmPFC. The dots represent the and its degree of purity was determined by the ratio of absor-
injection sites visualized by the dye injection. b Representative photomi-
crographs of the injection sites into the vmPFC bances measured at 260 and 280 nm (A260/A280). Values
between 1.75 and 2.0 were considered suitable for use. The
mRNA was reverse transcribed using a kit (#4374966,
LifeTechnologies), according to the manufacturer’s
Mol Neurobiol

instructions. DNA amplification reactions were run in tripli- [127]. Amplicon sequences of predesigned TaqMan gene ex-
cate using the TaqMan assays in StepOnePlus™Real-Time pression assays are provided in the Supplemental materials
PCR Systems (Applied Biosystems). The primers were (Table 1).
BDNF IV (forward primer: GGTGTAGGCTGGAA
TAGACTCTTG; reverse primer: GGAAAAGGATGGTC Drugs and Treatment
ATCACTCTTCT), TrkB (#Rn01441749_m1), 5-HT 1A
(#Rn00561409_s1), NMDA2a (#Rn00561341_m1), and 5-aza-2′-deoxycytidine (5-AzaD, nucleoside DNMT inhibi-
AMPA1 (#Rn00709588_m1). PCR conditions were 50 °C tor; Sigma-Aldrich, USA) was dissolved in sterile saline,
for 2 min (incubation) and 95 °C for 10 min (polymerase and N-phtalyl-L-tryptophan (RG108, non-nucleoside DNMT
activation), followed by 40 cycles of 95 °C for 15 s and inhibitor; Tocris Biosciences, UK) was diluted in 2% Tween
60 °C for 1 min (denaturation and annealing/extension). The 80 in sterile saline solution. Both drugs were intraperitoneally
mRNA expression levels were calculated using 2−ΔΔCT meth- administered at doses of 0.2, 0.4, and 0.6 mg/kg [102]. K252a
od. The internal standards were YWHAZ (TrkB receptor antagonist; Sigma-Aldrich, USA) was dis-
(#Rn00755072_m1), ACTB (#Rn00667869_m1), and solved in sterile saline and administered in a dose of
HPRT1 (#Rn01527840_m1) [126]. The results were normal- 10 pmol/0.2 μl/side [129]. Rapamycin (RAPA, mTOR
ized by the geometric mean of the reference genes, which blocker; LC Laboratories, USA) was dissolved in 0.2 μl
were previously verified for stability throughout treatment DMSO and administered in the dose of 0.2 nmol/0.2 μl/side
conditions. Amplicon sequences of predesigned TaqMan [65]. Imipramine hydrochloride (IMI, a tricyclic
gene expression assays are provided in the Supplemental ma- antidepressant; Sigma-Aldrich, USA) was dissolved in sterile
terials (Table 1). saline and injected i.p. at the dose of 15 mg/kg [101, 103].
Fluoxetine hydrochloride (a selective serotonin reuptake
DNA Methylation Analysis by meDIP and qPCR-RT inhibitor; Sigma-Aldrich, USA) was dissolved in 2% Tween
80 in sterile saline and injected i.p. at the dose of 20 mg/kg
For analyzing gene-specific DNA methylation changes, geno- [130]. All drugs were freshly prepared before use.
mic DNA was extracted using Wizard Genomic DNA
Purification kit (#A1120, Promega Corporation, USA), ac- Experimental Design
cording to the manufacturer’s protocol and sonicated (Sonics
Vibracell; 6 pulses, 20 s of duration, interval between pulses The studies were conducted as four independent experiments,
of 40 s and amplitude 40%) to random fragments of 200– all carried out between 8 AM and 2 PM. Animals were dis-
1000 bp [127]. One part of non-sonicated DNA was stored tributed to different treatment groups and experimental condi-
for control of sonication and an aliquot of DNA fragments tions in a randomized order to avoid potential circadian influ-
from each sample was stored for normalization of results in ences. The behavioral analysis was performed by experi-
the ratio of no-immunoprecipitated total DNA (input) [128]. menters that were blind to the different treatment conditions.
Immunoprecipitation of methylated DNA was performed
using a rabbit monoclonal 5-methylcytosine antibody Experiment 1: Antidepressant-Like Effects Induced by DNMTi
(5 μg/ml, #ab214727, Abcam) for 2 h at 4 °C. DNA was in the LH Paradigm
purified by Wizard Clean-up System kit (#A7280, Promega
Corporation, USA) according to the manufacturer’s instruc- To evaluate whether acute treatment with DNMTi would in-
tions. TaqMan probes were used for BDNF IV (forward prim- duce fast and/or sustained antidepressant effects in the LH,
er: GGTGTAGGCTGGAATAGACTCTTG; reverse primer: rats received an i.p. injections of 5-AzaD (0.6 mg/kg),
GGAAAAGGATGGTCATCACTCTTCT) and TrkB (#Rn RG108 (0.6 mg/kg), or vehicle (10 ml/kg), immediately after
04338713_g1). Immunoprecipitated (IP) and input DNA were the PT. Additional groups of animals were submitted to the PT
analyzed by qRT-PCR using TaqMan assays and received a systemic injection of the 5-AzaD (0.2 mg/kg),
(LifeTechnologies) and a StepOnePlus real-time PCR system RG108 (0.6 mg/kg), or vehicle 1 h before T (day 7). On day
(Applied Biosystems). PCR conditions were 50 °C for 2 min 7th, all animals were submitted to the T session (Fig. 3a). A
(incubation) and 95 °C for 10 min (polymerase activation), total of 122 animals were used in this experiment (5-AzaD and
followed by 40 cycles of 95 °C for 15 s and 60 °C for 1 min vehicle, 70 animals, Fig. 3b–d; RG108 and vehicle, 52 ani-
(denaturation and annealing/extension). YWHAZ mals, Fig. 3e–f).
(#Rn06427373_s1), ACTB (#Rn01412977_g1), and HPRT1 As a control experiment to demonstrate that the LH
(#Rn01318746_g1) were used as an internal standard. model is not sensitive to an acute intervention with a
Relative methylated DNA was calculated via the 2−ΔΔCt meth- conventional monoaminergic antidepressant, rats were
od and it was measured in triplicate. The gene-specific DNA submitted to the PT session and, immediately later
methylation levels were expressed by IP DNA/input DNA (day 1), received systemic injections of imipramine
Mol Neurobiol

(15 mg/kg), fluoxetine (20 mg/kg), or vehicle (10 ml/ Experiment 3: Changes in DNA Methylation in Response
kg). During the following 6 days, the animals received to RG108 Treatment in LH Animals
the administration of the drugs or vehicle and on day
7th, 1 h after the last injection, all animals were sub- To evaluate if the changes in gene expression levels were
mitted to the T session in the LH (Fig. 2a). The animals associated with changes in DNA methylation, rats were sub-
that received antidepressant drugs were divided in two jected to the PT in LH (day 1) and, immediately after PT (day
groups: (1) acute (ac), animals received 6 injections of 1) or 1 h before euthanasia (day 7), received i.p. injection of
the vehicle and only one injection of the drug on the RG108 (0.6 mg/kg) or vehicle (10 ml/kg). On day 7, the PFC
7th day; and (2) repeated (rep), animals received seven was dissected for molecular analysis using meDIP and qPCR-
injections of the drug. A total of 114 animals were used RT (Fig. 5a). An independent group of untreated and non-
in this experiment (imipramine and vehicle, 58 animals, stressed animals (naive control) was run in parallel for exper-
Fig. 2b–d; fluoxetine and vehicle, 56 animals, Fig. 2e– imental control. A total of 40 animals were used in this
g). experiment.

Experiment 2: DNMTi Effects on Gene Expression in the PFC Experiment 4: The Role of PFC BDNF-TrkB-mTOR Signaling
and HPC of LH Animals in the Antidepressant-Like Effect Induced by DNMTi

To evaluate if stress and/or DNMT inhibition could regulate To further evaluate the potential fast antidepressant ef-
the expression of relevant genes (5HT1A, TrkB, BDNF, fect of DNMTi, the effects of a single administration of
AMPA, NMDA), rats were subjected to the PT (day 1) and, DNMTi were investigated in rats subjected to the FST,
immediately after or 1 h before euthanasia (day 7), received which usually requires 3 administrations in 24 h [101].
i.p. injection of RG108 (0.6 mg/kg) or vehicle (10 ml/kg). On Rats were subjected to the FST (PT session; 15 min)
day 7, the brain regions (PFC, dorsal HPC, and ventral HPC) and immediately (0 h; Fig. 6a) or 23 h later (Fig. 6d)
were dissected for molecular analysis using qPCR-RT received intraperitoneal (i.p.) injections of DNMTi (5-
(Fig. 4a). An independent group of untreated and non- AzaD or RG108; 0.2 and 0.4 mg/kg) or vehicle
stressed animals (naive control) was run in parallel for exper- (10 ml/kg). As a positive control for the experiment,
imental control. A total of 40 animals were used in this independent groups of rats were subjected to the PT
experiment. and received three injections of imipramine (15 mg/kg),

Fig. 2 Antidepressant drugs attenuated the behavioral changes induced number of failures, and (d and g) did not change the number of
by footshocks stress. a Experimental scheme. b–d Imipramine (n = 9–10/ crossings. Veh, vehicle; ac, acute treatment; rep, repeated treatment. *
group). e–g Fluoxetine (n = 8–10/group). b and e Repeated antidepressant p < 0.05, rep vs veh stressed groups; # p < 0.05, stressed vs habituated
treatment increased the number of escapes, (c and f) decreased the groups
Mol Neurobiol

0 h, 5 h, and 23 h after the PT. Twenty-four hours after DNMTi Results in Fast and Sustained Antidepressant-
PT, the animals were subjected to the test (T; 5 min) Like Effects in LH
when the immobility time was measured. A total of 112
animals were used in this experiment (0 h, 5-AzaD, In order to evaluate possible fast antidepressant effects in-
imipramine, and vehicle: 38 animals, Fig. 6; RG108, duced by DNMTi in the LH, we first investigated if our be-
imipramine, and vehicle, 19 animals, Fig. 6c; 23 h, 5- havioral protocol was sensitive to the effect of antidepressants
AzaD, imipramine, and vehicle, 39 animals, Fig. 6e; only after repeated treatment, as described in the literature.
RG108, imipramine, and vehicle, 16 animals, Fig. 6f). Therefore, we assessed the effects of two different antidepres-
To evaluate if the BDNF-TrkB-mTOR signaling could be sants after acute and repeated treatment in animals exposed to
important for the antidepressant-like effect induced by LH.
DNMTi, rats were submitted to the PT and T, 24 h later. Our results revealed that a significant interaction between
One hour before the test, animal received intra-vmPFC bilat- factors was observed in all behavioral analysis (see Table 2 for
eral injections of rapamycin (RAPA, 0.2 nmol/0.2 μl/side), details); the effect of each treatment was analyzed separately
K252a (10 pmol/0.2 μl/side), or vehicle (0.2 μl/side) followed in stressed and in non-stressed groups. No effect of the drugs
by an i.p. injection of RG108 (0.2 mg/kg) or vehicle (10 ml/ was observed in the non-stressed groups, in any of the vari-
kg) (Fig. 6j). A total of 28 animals were used in this ables analyzed (p > 0.05, one-way ANOVA). However, in the
experiment. stressed groups, only the repeated treatment with imipramine
To investigate possible unspecific locomotor effects in- and fluoxetine attenuated the stress effects in the number of
duced by DNMTi, 5-AzaD (0.2 mg/kg), RG108 escapes (p < 0.05, Dunnets, Fig. 2b and e; Table 1) and in the
(0.2 mg/kg), or their vehicle (10 ml/kg) were administered number of failures (p < 0.05, Dunnets, Fig. 2c and f; Table 2).
intraperitoneally to independent groups of rats. The ani- These results indicate that only the repeated administration of
mals were submitted to the OFT 24 h (sustained effect) or conventional monoaminergic drugs can induce antidepressant
1 h (rapid effect) after the injection, and the number of effects in our protocol, with no observable effect of the single
crossings was measured for 5 min. Experimental scheme injection.
is shown in Fig. 6g. A total of 36 animals were used in Based on that, we next investigated the effects of a single
this experiment (5-AzaD and vehicle, 18 animals, Fig. 6h; administration of the two pharmacologically different
RG108 and vehicle, 18 animals, Fig. 6i). DNMTi, at two different time points. We observed that the
single administration of 5-AzaD increased the number of es-
Statistical Analyses capes (p < 0.05, Dunnets, Fig. 3b; Table 2) and decreased the
number of failures (p < 0.05, Dunnets, Fig. 3c; Table 2) in
All statistical analyses were performed with the Prism 7.0 stressed animals, without changing the behavior of the non-
software from GraphPad (La Jolla, CA, USA). Two-way anal- stressed ones. Similarly, RG108 attenuated the behavioral
ysis of variance (ANOVA) followed by Bonferroni’s test was changes induced by stress in LH by increasing the number
used to investigate the behavioral changes in the LH (factors, of escapes (p < 0.05, Dunnets, Fig. 3e; Table 2) and decreas-
treatment and condition). In the case of significant interaction ing the number of failures (p < 0.05, Dunnets, Fig. 3f;
between factors, specific post hoc comparisons in each condi- Table 2).
tion were performed with the help of the Dunnett’s test. The In all treatment conditions, no significant changes were
differences in mRNA expression and DNA methylation levels observed in the number of intertrial crossings (Figs. 2d, g,
between naïve and vehicle groups were analyzed by Student’s and 3d and g; Table 2). Altogether, the results indicate that
t test and 1-way ANOVA was used in stressed groups (vehicle the acute administration of pharmacologically different
and RG108 treatments). In FST and OFT, 1-way ANOVA DNMTi induces fast and sustained antidepressant effects in
followed by Dunnett’s test was performed. Differences were the LH.
regarded as significant if p < 0.05. F-values and degrees of
freedom are presented for ANOVAs, and t values are present- BDNF IV and TrkB Transcript Levels Are Reduced in
ed for Student’s t test. Data are shown as means ± standard Stressed Animals
error of means (S.E.M.). Animals that had injection site out-
side the vmPFC were excluded from the statistical analysis. Since both 5-AzaD and RG108 induced antidepressant effects
after a single i.p. administration, we next investigated if this
effect could be associated with changes in the expression level
Results of BDNF IV and TrkB, which are known to mediate the fast
antidepressant effect of other drugs. In the PFC, the mRNA
The statistical data are shown in Supplemental materials for BDNF IV and TrkB were significantly decreased in
(Tables 2–5). stressed rats (Fig. 4b–c; Table 3), but no significant stress-
Mol Neurobiol

Fig. 3 DNMTi induced rapid and sustained antidepressant-like effects in the number of failures, and (d and g) did not change the number of
rats submitted to the learned helplessness. a Experimental scheme. b–d 5- crossings. Veh, vehicle group. * p < 0.05, DNMTi treatments vs veh
AzaD (n = 11–12/group). e–g RG108 (n = 8–10/group). b and e Acute stressed groups; # p < 0.05, stressed vs habituated groups
DNMTi treatment increased the number of escapes, (c and f) decreased

Fig. 4 Footshocks stress reduced BDNF IV and TrkB mRNA levels in 9–11/group), (h) TrkB mRNA (n = 5–6/group), (i) 5-HT1A mRNA (n =
prefrontal cortex. a Experimental scheme. b–f Prefrontal cortex, PFC. g– 9–10/group), (j) NMDA2a mRNA (n = 9–11/group), and (k) AMPA1
k Dorsal hippocampus, dHPC. l–p Ventral hippocampus, vHPC. In PFC, mRNA (n = 9–11/group). Similarly, no alteration has been observed in
(b) BDNF IV mRNA (n = 9–10/group) and (c) mRNA TrkB (n = 8–10/ vHPC, (l) BDNF IV mRNA (n = 9–11/group), (m) TrkB mRNA (n = 8–
group) expressions were reduced by footshocks stress. None change was 10/group), (n) 5-HT1A mRNA (n = 8–11/group), (o) NMDA2a mRNA
observed in (d) 5-HT1A mRNA (n = 8–10/group), (e) NMDA2a mRNA (n = 8–11/group), and (p) AMPA1 (n = 8–11/group). Veh, vehicle. *
(n = 8–10/group), and (f) AMPA1 mRNA (n = 8–10/group) expressions. p < 0.05, vehicle vs naïve groups
No alteration also has been observed in dHPC, (g) BDNF IV mRNA (n =
Mol Neurobiol

induced changes were apparent in mRNA 5-HT1A (Fig. 4d; Discussion


Table 3) NMDA2a (Fig. 4e; Table 3) and AMPA1 (Fig. 4f;
Table 3). Interestingly, the levels of transcripts were not mod- The main findings in this work are that single administration
ified in RG108 treated rats when compared with the stressed of two distinct inhibitors of DNMT (5-AzaD and RG108)
control group (Fig. 4; Table 3). Moreover, no changes in tran- induces rapid and sustained antidepressant-like effects in
scripts levels were observed in response to stress or RG108 two different animal models, the LH and the FST.
treatment in both dorsal HPC (Fig. 4g–k; Table 2) and ventral Interestingly, intra-mPFC administration of a TrkB antagonist
HPC (Fig. 4l–p; Table 3). or an mTOR inhibitor abolished the antidepressant-like effect
induced by systemic administration of RG108. We also ob-
DNA Methylation Levels in BDNF IV and TrkB in served that treatment with RG108 attenuated stress-induced
Stressed Animals effects on DNA methylation in the expression of BDNF IV
and TrkB in the PFC. Collectively, these data suggest that the
To further investigate if the changes in the transcript levels antidepressant-like effects induced by DNMTi could involve
could reflect changes at the DNA methylation levels, we next rapid disinhibition of BDNF and TrkB expression, leading to
investigated stress and treatment effect on the methylation further downstream activation of mTOR in the PFC.
levels in BDNF IV and TrkB in the PFC. Results showed that Our findings are consistent with previous studies showing
stress increased and RG108 treatment attenuated DNA meth- that systemic administration of both RG108 and 5-AzaD has
ylation levels in BDNF IV and TrkB (p < 0.05; Fig. 5 b–c; the potential to induce an antidepressant-like effect in rats and
Table 4). mice subjected to the FST [101, 102]. However, here, we
show the first evidence that the acute administration of a
DNMTi Induces Rapid and Sustained Antidepressant- DNMTi promotes an antidepressant-like effect in the LH, a
Like Effects in the FST: Participation of TrkB-mTOR behavioral paradigm insensitive to acute administration of an-
Signaling tidepressant drugs [103, 131]. Earlier studies with intracere-
bral drug administration strongly suggest the involvement of
To further confirm the results observed in the LH test, we cortical and limbic brain regions in such effects, in that direct
investigated the effects of acute DNMTi administration in administration of DNMTi into the nucleus accumbens [104],
animals submitted to the FST. The results indicated that both the HPC [101], or ventrolateral orbital cortex [105] induces
5-AzaD and RG108, at both time points (immediately after PT significant antidepressant-like effects. Interestingly, previous
or 1 h before T), induced antidepressant-like effects in rats studies have shown that distinct subtypes of DNMT
submitted to the FST (p < 0.05; Fig. 6b–f, Table 5). None of (DNMT1, DNMT3a, and DNMT3b) are expressed in the
the treatments resulted in significant effects on the number of adult rodent HPC and PFC, where they play important roles
crossings in the OFT (Fig. 6h–i, Table 5). in transcription regulation of genes associated with synaptic
Administration of RAPA or K252a into the vmPFC plasticity, cognition, and behavioral responses to stress
prevented the antidepressant effect induced by systemic [132–135]. Moreover, in stressed animals, altered levels of
RG108 administration 1 h before the FST (Fig. 6k, Table 5), DNMT1 and DNMT3a have been described in the PFC and
thus indicating that BDNF-TrkB-mTOR activation is in- HPC of animals [103], suggesting that changes in DNMT
volved in the rapid antidepressant effect of DNMTi. levels and/or activity could contribute to aberrant DNA

Fig. 5 RG108 treatment attenuated BDNF IV and TrkB DNA BDNF IV, and (c) TrkB DNA methylation levels in PFC (n = 10/group).
methylation changes induced by stress. a Experimental scheme. The Veh, vehicle. * p < 0.0, veh vs naïve groups; # p < 0.05 RG108 vs vehicle
stress increased and the RG108 treatment attenuated this change in (b) groups
Mol Neurobiol

Fig. 6 DNMTi induced rapid and sustained antidepressant-like effect and (IMI; 3 injections). Imipramine treatment (3 injections) was used for
TrkB/mTOR inhibition blocked this behavioral effect induced by RG108 positive control. h and i No alteration has been indicated in the number
in forced swimming test. a, d, g, and j Experimental schemes. b, e, and h) of crossings (n = 6/group). k The pretreatment with rapamycin (RAPA) or
5-AzaD. c, f, and i RG108. Immediately or 23 h after the PT, (b and e) 5- K252a blocked the immobility time reduction induced by RG108 (n = 7/
AzaD (n = 9–11/group) and (c and f) RG108 (n = 4–5/group) administra- group). * p < 0.05, drug treatments vs vehicle or vehicle/vehicle
tion result in reduced immobility time in FST, similar to the imipramine

methylation and gene expression changes associated with analysis of biological material (saliva, blood, or post-mortem
stress and stress-related psychiatric disorders. Indeed, the brains) from depressed humans showed DNA methylation
Mol Neurobiol

changes compared with healthy controls [136–139]. injection of DNMTi. On the other hand, repeated administra-
Specifically, Poulter and coworkers (2008) demonstrated that tion with fluoxetine or imipramine was required to promote a
victims of suicide had altered levels of DNMT1 and DNMT3b significant antidepressant effect in the LH, leaving acute in-
in several brain regions, including cortex and amygdala, sug- jections ineffective. This evidence is in full agreement with
gesting alterations of both enzymes in the pathology of de- previously published papers indicating that the LH is not sen-
pressive disorder [136]. Interestingly, there is evidence that sitive to acute interventions with conventional monoaminergic
DNMT1 may play a more fundamental role than DNMT3, antidepressants [81, 103, 130], but it responds to single ad-
in that conditional forebrain deletion of DNMT1 induced an- ministration with ketamine, a fast-acting antidepressant drug
tidepressant and anxiolytic phenotype, whereas deletion of [146]. Therefore, the results observed in the LH suggest that
DNMT3a did not induce any behavioral changes [140]. In DNMT inhibition promotes fast antidepressant effects.
contrast, overexpression of DNMT3a in the nucleus accum- The mechanisms involved in this remarkable effect remain
bens and in the mPFC induced depressive-like and anxiogenic elusive and warrant further investigation. However, it can be
behavior in mice, respectively [141]. Accordingly, in a previ- proposed that inhibition of DNA methylation shortly after
ous study from our group, increased DNMT3a and DNMT3b stress exposure could favor changes that result in sustained
expression, along increased DNA methylation, was found in effects, as observed in this work. This could be attributed to
the PFC of helpless animals, an effect that was abolished by rapid and long-lasting changes in the expression of genes pro-
chronic, but not acute, antidepressant administration [103]. moting synaptic plasticity and adaptation to stress, since the
Interestingly, there are studies showing that repeated treat- drug is expected to be completely eliminated from the body
ment with antidepressants decreases DNMT levels and DNA when the behavioral testing takes place (time half-life of
methylation in the brain of animals exposed to different RG108, 4 h [147]; and 5-AzaD, 40 min [148]). Indeed, pre-
models of depression [103] and that clinical remission induced clinical works show that the behavioral effect of rapid-acting
by chronic antidepressant treatment is associated with normal- antidepressants involves an immediate increase in cortical [65,
ization of DNMT levels in blood cells of previously depressed 149, 150] and hippocampal [151–153] neuroplasticity.
patients [99]. It is therefore likely that the behavioral effect Ketamine is shown to rapidly increase the levels of BDNF
induced by chronic antidepressant treatment may be associat- and synaptic proteins in the HPC and PFC of animals exposed
ed with reversing aberrant DNA methylation in brain regions to stress models of depression [154]. As DNA methylation
involved in depression neurobiology. Therefore, 5-AzaD and regulates transcription of genes involved in neuroplasticity,
RG108 would trigger antidepressant effects by directly including BDNF and its receptor, TrkB, it can be hypothe-
targeting DNMTs and hence inhibiting stress-induced DNA sized that the antidepressant effect induced by RG108 could
methylation. be mediated by a decrease in DNA methylation of genes in-
Both RG108 and 5-AzaD are non-selective inhibitors of volved in synaptic plasticity with subsequent increase in their
DNMTs. 5-AzaD is a nucleoside analogue that, following transcript levels.
incorporation into DNA, traps DNMTs by covalent binding The expression levels of other genes in the HPC and PFC,
of the enzyme to DNA, blocking methyltransferase activity which have been shown to be regulated by DNA methylation
irreversibly [142]. RG108 is a non-nucleoside inhibitor that and are central to the antidepressant effect, were also investi-
binds to the active site of DNMT1, and presumably to gated herein, such as 5-HT1A, NMDA, and AMPA [78, 80,
DNMT3, and blocks the enzyme transiently [143–145]. The 92, 93, 106–109, 112, 115, 116, 155, 156]. However, we did
two chemically unrelated drugs are capable of inhibiting not detect any changes in the mRNA levels of 5-HT1A,
DNMTs by different mechanisms, providing greater assur- NMDA2a, and AMPA1, suggesting that they may not be di-
ance that the results observed with the administration of these rectly involved in the effects of DNMTi, or that our experi-
drugs would be related to inhibition of DNA methylation rath- mental design did not allow for the detection of such alter-
er than to unspecific mechanisms. Given the ability of both ations. Evaluation of DNA methylation and transcript levels at
drugs to bind to the catalytic domain of different DNMT sub- different time points after stress could provide additional im-
types, it is possible that the antidepressant effect observed portant information about that.
herein results from the inhibition of different DNMTs (1, 3a, The main finding in our work is that exposure to
and 3b) and subsequent disinhibition of gene expression in footshocks in the LH paradigm increased DNA methylation
different brain regions involved in depression neurobiology. and reduced mRNA levels of BDNF exon IV and TrkB in the
However, future studies should test selective inhibitors to as- PFC. This is in line with previous evidence showing that LH
sess the specific contribution of each DNMT subtype to the exposure increased global DNA methylation and DNMT3
effects observed in this work. expression in the PFC [103]. RG108 treatment attenuated
A key finding here is that acute intervention with DNMTi BDNF and TrkB DNA methylation changes induced by stress
promotes fast antidepressant effects, observed as a significant but failed to affect BDNF and TrkB transcript levels.
attenuation of escape failures in animals receiving a single However, inhibition of mTOR, as well as TrkB antagonism,
Mol Neurobiol

blocked the rapid antidepressant-like effect induced by acute not measure mRNA levels at other time points, nor protein
RG108 treatment in the rat FST, thus suggesting that BDNF- levels at the moment when behavioral changes were observed.
TrkB-mTOR signaling in the PFC is involved in the As we chose to euthanize the animals at the moment that they
antidepressant-like behavior induced by RG108, but in a man- would have been exposed to the test in the LH, we were
ner which cannot be detected on the transcript levels. Further unable to evaluate how the molecular changes would correlate
studies are required to in-depth determine the mechanisms with the behavior. Such studies should be performed in future
involved. Again, it is possible that changes at different time work, as it could reveal important differences in DNA meth-
points might have occurred which have not been investigated ylation and transcript levels in animals that are susceptible and
in the present study. resilient to stress, and well as animals that respond and do not
BDNF is a neurotrophin widely expressed in the CNS as- respond to antidepressants.
sociated with cell survival and differentiation, organization, It is also important to consider that changes in DNA meth-
and maintenance of neural activity, and it is closely associated ylation levels do not always correspond to changes in gene
with the neurobiology of depression [12, 93, 157–159]. expression, since other steps in the expression-process (for
Relevant to the present work, it has been shown that stress example splicing and translation) may also be regulated epi-
exposure can alter DNA methylation levels and modulate genetically [166]. Indeed, recent evidence indicates that DNA
BDNF expression in different brain structures. For example, methylation can also be functionally silent, i.e., that it does not
rats subjected to reduced maternal care have increased DNA necessarily result in equivalent changes in gene expression
methylation levels in BDNF IV accompanied by reduced [167]. Furthermore, rapid demethylation and de novo methyl-
levels of this protein in the PFC [160]. Activation of the ation after any given treatment intervention can impair the
BDNF main receptor, TrkB, activates intracellular signaling observation of changes in DNA methylation levels and sub-
cascades including mitogen-activated protein kinases/ sequent gene expression changes in each moment [167]. Of
extracellular signal-related kinases (MAPK/ERK), phospholi- note, the assays performed in the intra exon region may not
pase Cγ (PLCγ1)/PKC, and the phosphatidyl inositol kinase correlate with DNA methylation in the promoter region of
(PI3K)/AKT/mTOR. Administration of a TrkB agonist, 7,8 genes neither with corresponding gene transcription changes.
dihydroflavone (7,8 DHF) can induce an antidepressant-like The lack of more details on the gene sequence analyzed due to
effect in mice exposed to the FST and TST and increase PSD- the use of predesigned assays also makes the in-depth inter-
95 (postsynaptic density protein 95) levels, a marker of syn- pretation of the results more difficult, thus remaining as a
aptogenesis, in the PFC and HPC of socially defeated mice limitation of the present study.
[161]. Also, bilateral 7,8 DHF infusion into the medial PFC In conclusion, our pharmacological data provides
(infralimbic) induces an antidepressant-like effect in rats sub- strong evidence that acute intervention with inhibitors
mitted to LH, suggesting that the activation of TrkB receptor of DNA methylation attenuates stress-induced DNA
in the PFC triggers behavioral adaptation to stress and antide- methylation on BDNF and TrkB in the PFC and elicits
pressant effect [162]. Intriguingly, DNA methylation in the rapid antidepressant-like effects. Furthermore, BDNF-
TrkB gene has been linked to the effects of stress, and studies TrkB signaling in the PFC is required for the
of the cortex of suicidal individuals demonstrated increased antidepressant-like effect induced by DNMT inhibition.
DNA methylation accompanied by reduced TrkB expression Our data provide convincing evidence that inhibition of
[163], further substantiating the role of BDNF-TrkB pathway DNA methylation could constitute a novel approach to
as crucial in depression. rapid and sustained antidepressant effects.
In addition to DNA methylation, mRNA levels can also be
acutely modulated during stress response [164]. However, in
Acknowledgments The authors acknowledge Prof. Francisco Silveira
this work, we failed to attenuate stress effects on BDNF and Guimarães for his helpful contribution to the statistical analysis and
TrkB mRNA levels in the rat PFC using RG108 treatment. Flávia Fiacadori Salata for her technical assistance. We are thankful to
Several possible explanations exist. Since the design of the Prof. Gregers Wegener for his comments and suggestions on the
study was cross-sectional and not longitudinal, it is possible manuscript.
that the time of analysis does not coincide with the ideal mo-
Authors’ Contributions A.J.S. performed experiments, analyzed data,
ment for identifying changes in mRNA levels. For example, it and wrote the paper. I.S.M. and AC.D.R.S. performed experiments.
has been shown that mRNA changes in rats chronically S.R.L.J. designed the experiments, supervised the project, and contribut-
stressed for 21 days are absent, while decreased protein ex- ed with writing the paper. All authors discussed the results, implications,
pression of NMDA2a and NMDA2b (24 h after stress) in the and commented on the manuscript at all stages.
frontal cortex could still be detected [165]. Therefore, it can be
Funding This work was supported by research grants from the Research
speculated that changes in mRNA levels at another time point Foundation of the State of São Paulo (FAPESP, grant numbers: S.R.L.J.,
could have happened, but remained undetected in our work. In 2011/17281-7; 2012/17626-7; A.J.S., 2015/01955-0), CNPQ (304780/
this context, it is a limitation of the present study that we did 2018-9; 06648/2014-8).
Mol Neurobiol

Compliance with Ethical Standards precursor BDNF to CNS neurons: negative regulation of neurite
growth, spine formation and cell survival. Mol Brain 2:27. https://
doi.org/10.1186/1756-6606-2-27
Conflict of Interest The authors declare that they have no competing
16. Woo NH, Teng HK, Siao CJ, Chiaruttini C, Pang PT, Milner TA,
interests.
Hempstead BL, Lu B (2005) Activation of p75NTR by proBDNF
facilitates hippocampal long-term depression. Nat Neurosci 8(8):
1069–1077. https://doi.org/10.1038/nn1510
References 17. Sen S, Duman R, Sanacora G (2008) Serum brain-derived neuro-
trophic factor, depression, and antidepressant medications: meta-
analyses and implications. Biol Psychiatry 64(6):527–532. https://
1. Vigo D, Thornicroft G, Atun R (2016) Estimating the true global
doi.org/10.1016/j.biopsych.2008.05.005
burden of mental illness. Lancet Psychiatry 3(2):171–178. https://
18. Shimizu E, Hashimoto K, Okamura N, Koike K, Komatsu N,
doi.org/10.1016/S2215-0366(15)00505-2
Kumakiri C, Nakazato M, Watanabe H et al (2003) Alterations
2. Pittenger C, Duman RS (2008) Stress, depression, and
of serum levels of brain-derived neurotrophic factor (BDNF) in
neuroplasticity: a convergence of mechanisms.
depressed patients with or without antidepressants. Biol
Neuropsychopharmacology 33(1):88–109. https://doi.org/10.
Psychiatry 54(1):70–75. https://doi.org/10.1016/s0006-3223(03)
1038/sj.npp.1301574
00181-1
3. American Psychiatric Association (2013) Diagnostic and statisti-
19. Koponen E, Lakso M, Castren E (2004) Overexpression of the
cal manual of mental disorders, Fifth Edn. Arlington, VA
full-length neurotrophin receptor trkB regulates the expression of
4. Shadrina M, Bondarenko EA, Slominsky PA (2018) Genetics plasticity-related genes in mouse brain. Brain Res Mol Brain Res
factors in major depression disease. Front Psychiatry 9:334. 130(1–2):81–94. https://doi.org/10.1016/j.molbrainres.2004.07.
https://doi.org/10.3389/fpsyt.2018.00334 010
5. Yang T, Nie Z, Shu H, Kuang Y, Chen X, Cheng J, Yu S, Liu H 20. Rantamaki T, Hendolin P, Kankaanpaa A, Mijatovic J, Piepponen
(2020) The role of BDNF on neural plasticity in depression. Front P, Domenici E, Chao MV, Mannisto PT et al (2007)
Cell Neurosci 14:82. https://doi.org/10.3389/fncel.2020.00082 Pharmacologically diverse antidepressants rapidly activate brain-
6. Dunham JS, Deakin JF, Miyajima F, Payton A, Toro CT (2009) derived neurotrophic factor receptor TrkB and induce
Expression of hippocampal brain-derived neurotrophic factor and phospholipase-Cgamma signaling pathways in mouse brain.
its receptors in Stanley consortium brains. J Psychiatr Res 43(14): Neuropsychopharmacology 32(10):2152–2162. https://doi.org/
1175–1184. https://doi.org/10.1016/j.jpsychires.2009.03.008 10.1038/sj.npp.1301345
7. Emon MPZ, Das R, Nishuty NL, Shalahuddin Qusar MMA, 21. Saarelainen T, Hendolin P, Lucas G, Koponen E, Sairanen M,
Bhuiyan MA, Islam MR (2020) Reduced serum BDNF levels MacDonald E, Agerman K, Haapasalo A et al (2003) Activation
are associated with the increased risk for developing MDD: a of the TrkB neurotrophin receptor is induced by antidepressant
case-control study with or without antidepressant therapy. BMC drugs and is required for antidepressant-induced behavioral ef-
Res Notes 13(1):83. https://doi.org/10.1186/s13104-020-04952-3 fects. J Neurosci 23(1):349–357
8. Guilloux JP, Douillard-Guilloux G, Kota R, Wang X, Gardier 22. Shirayama Y, Chen AC, Nakagawa S, Russell DS, Duman RS
AM, Martinowich K, Tseng GC, Lewis DA et al (2012) (2002) Brain-derived neurotrophic factor produces antidepressant
Molecular evidence for BDNF- and GABA-related dysfunctions effects in behavioral models of depression. J Neurosci 22(8):
in the amygdala of female subjects with major depression. Mol 3251–3261 20026292
Psychiatry 17(11):1130–1142. https://doi.org/10.1038/mp.2011. 23. Ye Y, Wang G, Wang H, Wang X (2011) Brain-derived neuro-
113 trophic factor (BDNF) infusion restored astrocytic plasticity in the
9. Thompson Ray M, Weickert CS, Wyatt E, Webster MJ (2011) hippocampus of a rat model of depression. Neurosci Lett 503(1):
Decreased BDNF, trkB-TK+ and GAD67 mRNA expression in 15–19. https://doi.org/10.1016/j.neulet.2011.07.055
the hippocampus of individuals with schizophrenia and mood dis- 24. Altar CA, Whitehead RE, Chen R, Wortwein G, Madsen TM
orders. J Psychiatry Neurosci 36(3):195–203. https://doi.org/10. (2003) Effects of electroconvulsive seizures and antidepressant
1503/jpn.100048 drugs on brain-derived neurotrophic factor protein in rat brain.
10. Tripp A, Oh H, Guilloux JP, Martinowich K, Lewis DA, Sibille E Biol Psychiatry 54(7):703–709. https://doi.org/10.1016/s0006-
(2012) Brain-derived neurotrophic factor signaling and subgenual 3223(03)00073-8
anterior cingulate cortex dysfunction in major depressive disorder. 25. Autry AE, Monteggia LM (2012) Brain-derived neurotrophic fac-
Am J Psychiatry 169(11):1194–1202. https://doi.org/10.1176/ tor and neuropsychiatric disorders. Pharmacol Rev 64(2):238–
appi.ajp.2012.12020248 258. https://doi.org/10.1124/pr.111.005108
11. Duman RS (2002) Pathophysiology of depression: the concept of 26. Molteni R, Calabrese F, Cattaneo A, Mancini M, Gennarelli M,
synaptic plasticity. Eur Psychiatry 17(Suppl 3):306–310. https:// Racagni G, Riva MA (2009) Acute stress responsiveness of the
doi.org/10.1016/s0924-9338(02)00654-5 neurotrophin BDNF in the rat hippocampus is modulated by
12. Duman RS, Monteggia LM (2006) A neurotrophic model for chronic treatment with the antidepressant duloxetine.
stress-related mood disorders. Biol Psychiatry 59(12):1116– Neuropsychopharmacology 34(6):1523–1532. https://doi.org/10.
1127. https://doi.org/10.1016/j.biopsych.2006.02.013 1038/npp.2008.208
13. Smith MA, Makino S, Kvetnansky R, Post RM (1995) Stress and 27. Nibuya M, Nestler EJ, Duman RS (1996) Chronic antidepressant
glucocorticoids affect the expression of brain-derived neurotroph- administration increases the expression of cAMP response ele-
ic factor and neurotrophin-3 mRNAs in the hippocampus. J ment binding protein (CREB) in rat hippocampus. J Neurosci
Neurosci 15(3 Pt 1):1768–1777 16(7):2365–2372
14. Taliaz D, Stall N, Dar DE, Zangen A (2010) Knockdown of brain- 28. Russo-Neustadt A, Beard RC, Cotman CW (1999) Exercise, an-
derived neurotrophic factor in specific brain sites precipitates be- tidepressant medications, and enhanced brain derived neurotroph-
haviors associated with depression and reduces neurogenesis. Mol ic factor expression. Neuropsychopharmacology 21(5):679–682.
Psychiatry 15(1):80–92. https://doi.org/10.1038/mp.2009.67 https://doi.org/10.1016/S0893-133X(99)00059-7
15. Koshimizu H, Kiyosue K, Hara T, Hazama S, Suzuki S, Uegaki K, 29. Russo-Neustadt AA, Alejandre H, Garcia C, Ivy AS, Chen MJ
Nagappan G, Zaitsev E et al (2009) Multiple functions of (2004) Hippocampal brain-derived neurotrophic factor expression
Mol Neurobiol

following treatment with reboxetine, citalopram, and physical ex- glutamate, alanine and serine with severity of depression. Prog
ercise. Neuropsychopharmacology 29(12):2189–2199. https://doi. Neuro-Psychopharmacol Biol Psychiatry 30(6):1155–1158.
org/10.1038/sj.npp.1300514 https://doi.org/10.1016/j.pnpbp.2006.03.036
30. Castren E, Rantamaki T (2010) The role of BDNF and its recep- 45. Naughton M, Clarke G, O’Leary OF, Cryan JF, Dinan TG (2014)
tors in depression and antidepressant drug action: reactivation of A review of ketamine in affective disorders: current evidence of
developmental plasticity. Dev Neurobiol 70(5):289–297. https:// clinical efficacy, limitations of use and pre-clinical evidence on
doi.org/10.1002/dneu.20758 proposed mechanisms of action. J Affect Disord 156:24–35.
31. Castren E, Rantamaki T (2010) Role of brain-derived neurotroph- https://doi.org/10.1016/j.jad.2013.11.014
ic factor in the aetiology of depression: implications for pharma- 46. Bartanusz V, Aubry JM, Pagliusi S, Jezova D, Baffi J, Kiss JZ
cological treatment. CNS Drugs 24(1):1–7. https://doi.org/10. (1995) Stress-induced changes in messenger RNA levels of N-
2165/11530010-000000000-00000 methyl-D-aspartate and AMPA receptor subunits in selected re-
32. Siuciak JA, Boylan C, Fritsche M, Altar CA, Lindsay RM (1996) gions of the rat hippocampus and hypothalamus. Neuroscience
BDNF increases monoaminergic activity in rat brain following 66(2):247–252. https://doi.org/10.1016/0306-4522(95)00084-v
intracerebroventricular or intraparenchymal administration. Brain 47. Fitzgerald LW, Ortiz J, Hamedani AG, Nestler EJ (1996) Drugs of
Res 710(1–2):11–20. https://doi.org/10.1016/0006-8993(95) abuse and stress increase the expression of GluR1 and NMDAR1
01289-3 glutamate receptor subunits in the rat ventral tegmental area: com-
33. Wong YH, Lee CM, Xie W, Cui B, Poo MM (2015) Activity- mon adaptations among cross-sensitizing agents. J Neurosci
dependent BDNF release via endocytic pathways is regulated by 16(1):274–282
synaptotagmin-6 and complexin. Proc Natl Acad Sci U S A 48. Marsden WN (2011) Stressor-induced NMDAR dysfunction as a
112(32):E4475–E4484. https://doi.org/10.1073/pnas.1511830112 unifying hypothesis for the aetiology, pathogenesis and comorbid-
34. Sakata K, Martinowich K, Woo NH, Schloesser RJ, Jimenez DV, ity of clinical depression. Med Hypotheses 77(4):508–528. https://
Ji Y, Shen L, Lu B (2013) Role of activity-dependent BDNF doi.org/10.1016/j.mehy.2011.06.021
expression in hippocampal-prefrontal cortical regulation of behav- 49. Masrour FF, Peeri M, Azarbayjani MA, Hosseini MJ (2018)
ioral perseverance. Proc Natl Acad Sci U S A 110(37):15103– Voluntary exercise during adolescence mitigated negative the ef-
15108. https://doi.org/10.1073/pnas.1222872110 fects of maternal separation stress on the depressive-like behaviors
35. Altamura CA, Mauri MC, Ferrara A, Moro AR, D’Andrea G, of adult male rats: role of NMDA receptors. Neurochem Res
Zamberlan F (1993) Plasma and platelet excitatory amino acids 43(5):1067–1074. https://doi.org/10.1007/s11064-018-2519-6
in psychiatric disorders. Am J Psychiatry 150(11):1731–1733.
50. McCarthy DJ, Alexander R, Smith MA, Pathak S, Kanes S, Lee
https://doi.org/10.1176/ajp.150.11.1731
CM, Sanacora G (2012) Glutamate-based depression GBD. Med
36. Deutschenbaur L, Beck J, Kiyhankhadiv A, Muhlhauser M,
Hypotheses 78(5):675–681. https://doi.org/10.1016/j.mehy.2012.
Borgwardt S, Walter M, Hasler G, Sollberger D et al (2016)
02.009
Role of calcium, glutamate and NMDA in major depression and
51. Moghaddam B (1993) Stress preferentially increases
therapeutic application. Prog Neuro-Psychopharmacol Biol
extraneuronal levels of excitatory amino acids in the prefrontal
Psychiatry 64:325–333. https://doi.org/10.1016/j.pnpbp.2015.02.
cortex: comparison to hippocampus and basal ganglia. J
015
Neurochem 60(5):1650–1657. https://doi.org/10.1111/j.1471-
37. Frye MA, Tsai GE, Huggins T, Coyle JT, Post RM (2007) Low
4159.1993.tb13387.x
cerebrospinal fluid glutamate and glycine in refractory affective
52. Sathyanesan M, Haiar JM, Watt MJ, Newton SS (2017) Restraint
disorder. Biol Psychiatry 61(2):162–166. https://doi.org/10.1016/
stress differentially regulates inflammation and glutamate receptor
j.biopsych.2006.01.024
gene expression in the hippocampus of C57BL/6 and BALB/c
38. Ghasemi M, Phillips C, Trillo L, De Miguel Z, Das D, Salehi A
mice. Stress 20(2):197–204. https://doi.org/10.1080/10253890.
(2014) The role of NMDA receptors in the pathophysiology and
2017.1298587
treatment of mood disorders. Neurosci Biobehav Rev 47:336–
358. https://doi.org/10.1016/j.neubiorev.2014.08.017 53. Weiland NG, Orchinik M, Tanapat P (1997) Chronic corticoste-
39. Hashimoto K, Sawa A, Iyo M (2007) Increased levels of gluta- rone treatment induces parallel changes in N-methyl-D-aspartate
mate in brains from patients with mood disorders. Biol Psychiatry receptor subunit messenger RNA levels and antagonist binding
62(11):1310–1316. https://doi.org/10.1016/j.biopsych.2007.03. sites in the hippocampus. Neuroscience 78(3):653–662. https://
017 doi.org/10.1016/s0306-4522(96)00619-7
40. Kim JS, Schmid-Burgk W, Claus D, Kornhuber HH (1982) 54. Jaso BA, Niciu MJ, Iadarola ND, Lally N, Richards EM, Park M,
Increased serum glutamate in depressed patients. Archiv Ballard ED, Nugent AC et al (2017) Therapeutic modulation of
Psychiat Nervenkrankheiten 232(4):299–304. https://doi.org/10. glutamate receptors in major depressive disorder. Curr
1007/BF00345492 Neuropharmacol 15(1):57–70. https://doi.org/10.2174/
41. Levine J, Panchalingam K, Rapoport A, Gershon S, McClure RJ, 1570159x14666160321123221
Pettegrew JW (2000) Increased cerebrospinal fluid glutamine 55. Berman RM, Cappiello A, Anand A, Oren DA, Heninger GR,
levels in depressed patients. Biol Psychiatry 47(7):586–593. Charney DS, Krystal JH (2000) Antidepressant effects of keta-
https://doi.org/10.1016/s0006-3223(99)00284-x mine in depressed patients. Biol Psychiatry 47(4):351–354.
42. Mathews DC, Henter ID, Zarate CA (2012) Targeting the gluta- https://doi.org/10.1016/s0006-3223(99)00230-9
matergic system to treat major depressive disorder: rationale and 56. Trullas R, Skolnick P (1990) Functional antagonists at the NMDA
progress to date. Drugs 72(10):1313–1333. https://doi.org/10. receptor complex exhibit antidepressant actions. Eur J Pharmacol
2165/11633130-000000000-00000 185(1):1–10. https://doi.org/10.1016/0014-2999(90)90204-j
43. Mauri MC, Ferrara A, Boscati L, Bravin S, Zamberlan F, Alecci 57. Zarate CA Jr, Singh JB, Carlson PJ, Brutsche NE, Ameli R,
M, Invernizzi G (1998) Plasma and platelet amino acid concentra- Luckenbaugh DA, Charney DS, Manji HK (2006) A randomized
tions in patients affected by major depression and under trial of an N-methyl-D-aspartate antagonist in treatment-resistant
fluvoxamine treatment. Neuropsychobiology 37(3):124–129. major depression. Arch Gen Psychiatry 63(8):856–864. https://
https://doi.org/10.1159/000026491 doi.org/10.1001/archpsyc.63.8.856
44. Mitani H, Shirayama Y, Yamada T, Maeda K, Ashby CR Jr, 58. Krystal JH, Sanacora G, Duman RS (2013) Rapid-acting gluta-
Kawahara R (2006) Correlation between plasma levels of matergic antidepressants: the path to ketamine and beyond. Biol
Mol Neurobiol

Psychiatry 73(12):1133–1141. https://doi.org/10.1016/j.biopsych. 73. Duric V, Banasr M, Stockmeier CA, Simen AA, Newton SS,
2013.03.026 Overholser JC, Jurjus GJ, Dieter L et al (2013) Altered expression
59. Du J, Suzuki K, Wei Y, Wang Y, Blumenthal R, Chen Z, Falke C, of synapse and glutamate related genes in post-mortem hippocam-
Zarate CA Jr et al (2007) The anticonvulsants lamotrigine, pus of depressed subjects. Int J Neuropsychopharmacol 16(1):69–
riluzole, and valproate differentially regulate AMPA receptor 82. https://doi.org/10.1017/S1461145712000016
membrane localization: relationship to clinical effects in mood 74. Toth E, Gersner R, Wilf-Yarkoni A, Raizel H, Dar DE, Richter-
disorders. Neuropsychopharmacology 32(4):793–802. https:// Levin G, Levit O, Zangen A (2008) Age-dependent effects of
doi.org/10.1038/sj.npp.1301178 chronic stress on brain plasticity and depressive behavior. J
60. Freudenberg F, Celikel T, Reif A (2015) The role of alpha-amino- Neurochem 107(2):522–532. https://doi.org/10.1111/j.1471-
3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) recep- 4159.2008.05642.x
tors in depression: central mediators of pathophysiology and anti- 75. Ferrari F, Villa RF (2017) The neurobiology of depression: an
depressant activity? Neurosci Biobehav Rev 52:193–206. https:// integrated overview from biological theories to clinical evidence.
doi.org/10.1016/j.neubiorev.2015.03.005 Mol Neurobiol 54(7):4847–4865. https://doi.org/10.1007/s12035-
61. Aan Het Rot M, Zarate CA Jr, Charney DS, Mathew SJ (2012) 016-0032-y
Ketamine for depression: where do we go from here? Biol 76. Kaufman J, DeLorenzo C, Choudhury S, Parsey RV (2016) The 5-
Psychiatry 72(7):537–547. https://doi.org/10.1016/j.biopsych. HT1A receptor in major depressive disorder. Eur
2012.05.003 Neuropsychopharmacol 26(3):397–410. https://doi.org/10.1016/
62. Ionescu DF, Swee MB, Pavone KJ, Taylor N, Akeju O, Baer L, j.euroneuro.2015.12.039
Nyer M, Cassano P et al (2016) Rapid and sustained reductions in 77. Yohn CN, Gergues MM, Samuels BA (2017) The role of 5-HT
current suicidal ideation following repeated doses of intravenous receptors in depression. Mol Brain 10(1):28. https://doi.org/10.
ketamine: secondary analysis of an open-label study. J Clin 1186/s13041-017-0306-y
Psychiatry 77(6):e719–e725. https://doi.org/10.4088/JCP. 78. Cheetham SC, Crompton MR, Katona CL, Horton RW (1990)
15m10056 Brain 5-HT1 binding sites in depressed suicides.
63. Price RB, Mathew SJ (2015) Does ketamine have anti-suicidal Psychopharmacology 102(4):544–548. https://doi.org/10.1007/
properties? Current status and future directions. CNS Drugs bf02247138
29(3):181–188. https://doi.org/10.1007/s40263-015-0232-4
79. Stockmeier CA, Shapiro LA, Dilley GE, Kolli TN, Friedman L,
64. Koike H, Fukumoto K, Iijima M, Chaki S (2013) Role of BDNF/
Rajkowska G (1998) Increase in serotonin-1A autoreceptors in the
TrkB signaling in antidepressant-like effects of a group II metab-
midbrain of suicide victims with major depression-postmortem
otropic glutamate receptor antagonist in animal models of depres-
evidence for decreased serotonin activity. J Neurosci 18(18):
sion. Behav Brain Res 238:48–52. https://doi.org/10.1016/j.bbr.
7394–7401
2012.10.023
80. Carr GV, Lucki I (2011) The role of serotonin receptor subtypes in
65. Li N, Lee B, Liu RJ, Banasr M, Dwyer JM, Iwata M, Li XY,
treating depression: a review of animal studies.
Aghajanian G et al (2010) mTOR-dependent synapse formation
Psychopharmacology 213(2–3):265–287. https://doi.org/10.
underlies the rapid antidepressant effects of NMDA antagonists.
1007/s00213-010-2097-z
Science 329(5994):959–964. https://doi.org/10.1126/science.
81. Joca SR, Padovan CM, Guimaraes FS (2003) Activation of post-
1190287
synaptic 5-HT(1A) receptors in the dorsal hippocampus prevents
66. Liu RJ, Lee FS, Li XY, Bambico F, Duman RS, Aghajanian GK
learned helplessness development. Brain Res 978(1–2):177–184.
(2012) Brain-derived neurotrophic factor Val66Met allele impairs
https://doi.org/10.1016/s0006-8993(03)02943-3
basal and ketamine-stimulated synaptogenesis in prefrontal cortex.
Biol Psychiatry 71(11):996–1005. https://doi.org/10.1016/j. 82. Pitchot W, Hansenne M, Pinto E, Reggers J, Fuchs S, Ansseau M
biopsych.2011.09.030 (2005) 5-Hydroxytryptamine 1A receptors, major depression, and
67. Autry AE, Adachi M, Nosyreva E, Na ES, Los MF, Cheng PF, suicidal behavior. Biol Psychiatry 58(11):854–858. https://doi.
Kavalali ET, Monteggia LM (2011) NMDA receptor blockade at org/10.1016/j.biopsych.2005.05.042
rest triggers rapid behavioural antidepressant responses. Nature 83. Savitz JB, Drevets WC (2013) Neuroreceptor imaging in depres-
475(7354):91–95. https://doi.org/10.1038/nature10130 sion. Neurobiol Dis 52:49–65. https://doi.org/10.1016/j.nbd.2012.
68. Maeng S, Zarate CA Jr, Du J, Schloesser RJ, McCammon J, Chen 06.001
G, Manji HK (2008) Cellular mechanisms underlying the antide- 84. Duman RS, Heninger GR, Nestler EJ (1997) A molecular and
pressant effects of ketamine: role of alpha-amino-3-hydroxy-5- cellular theory of depression. Arch Gen Psychiatry 54(7):597–606
methylisoxazole-4-propionic acid receptors. Biol Psychiatry 85. Depoortere R, Papp M, Gruca P, Lason-Tyburkiewicz M,
63(4):349–352. https://doi.org/10.1016/j.biopsych.2007.05.028 Niemczyk M, Varney MA, Newman-Tancredi A (2019)
69. Hashimoto K (2011) Role of the mTOR signaling pathway in the Cortical 5-hydroxytryptamine 1A receptor biased agonist, NLX-
rapid antidepressant action of ketamine. Expert Rev Neurother 101, displays rapid-acting antidepressant-like properties in the rat
11(1):33–36. https://doi.org/10.1586/ern.10.176 chronic mild stress model. J Psychopharmacol 33(11):1456–
70. Koike H, Iijima M, Chaki S (2011) Involvement of AMPA recep- 1466. https://doi.org/10.1177/0269881119860666
tor in both the rapid and sustained antidepressant-like effects of 86. Newman-Tancredi A, Bardin L, Auclair A, Colpaert F,
ketamine in animal models of depression. Behav Brain Res Depoortere R, Varney MA (2018) NLX-112, a highly selective
224(1):107–111. https://doi.org/10.1016/j.bbr.2011.05.035 5-HT1A receptor agonist, mediates analgesia and antidepressant-
71. Luscher B, Feng M, Jefferson SJ (2020) Antidepressant mecha- like activity in rats via spinal cord and prefrontal cortex 5-HT1A
nisms of ketamine: focus on GABAergic inhibition. Adv receptors, respectively. Brain Res 1688:1–7. https://doi.org/10.
Pharmacol 89:43–78. https://doi.org/10.1016/bs.apha.2020.03. 1016/j.brainres.2018.03.016
002 87. Ago Y, Tanabe W, Higuchi M, Tsukada S, Tanaka T, Yamaguchi
72. Beneyto M, Kristiansen LV, Oni-Orisan A, McCullumsmith RE, T, Igarashi H, Yokoyama R et al (2019) (R)-ketamine induces a
Meador-Woodruff JH (2007) Abnormal glutamate receptor ex- greater increase in prefrontal 5-HT release than (S)-ketamine and
pression in the medial temporal lobe in schizophrenia and mood ketamine metabolites via an AMPA receptor-independent mecha-
disorders. Neuropsychopharmacology 32(9):1888–1902. https:// nism. Int J Neuropsychopharmacol 22(10):665–674. https://doi.
doi.org/10.1038/sj.npp.1301312 org/10.1093/ijnp/pyz041
Mol Neurobiol

88. Pham TH, Mendez-David I, Defaix C, Guiard BP, Tritschler L, DNA methylation levels. Acta Neuropsychiatrica 28(1):11–22.
David DJ, Gardier AM (2017) Ketamine treatment involves me- https://doi.org/10.1017/neu.2015.40
dial prefrontal cortex serotonin to induce a rapid antidepressant- 103. Sales AJ, Joca SRL (2018) Antidepressant administration modu-
like activity in BALB/cJ mice. Neuropharmacology 112(Pt A): lates stress-induced DNA methylation and DNA methyltransfer-
198–209. https://doi.org/10.1016/j.neuropharm.2016.05.010 ase expression in rat prefrontal cortex and hippocampus. Behav
89. Jin HJ, Pei L, Li YN, Zheng H, Yang S, Wan Y, Mao L, Xia YP Brain Res 343:8–15. https://doi.org/10.1016/j.bbr.2018.01.022
et al (2017) Alleviative effects of fluoxetine on depressive-like 104. LaPlant Q, Vialou V, Covington HE 3rd, Dumitriu D, Feng J,
behaviors by epigenetic regulation of BDNF gene transcription Warren BL, Maze I, Dietz DM et al (2010) Dnmt3a regulates
in mouse model of post-stroke depression. Sci Rep 7(1):14926. emotional behavior and spine plasticity in the nucleus accumbens.
https://doi.org/10.1038/s41598-017-13929-5 Nat Neurosci 13(9):1137–1143. https://doi.org/10.1038/nn.2619
90. Takeuchi N, Nonen S, Kato M, Wakeno M, Takekita Y, Kinoshita 105. Xing B, Liu P, Xu WJ, Xu FY, Dang YH (2014) Effect of
T, Kugawa F (2017) Therapeutic response to paroxetine in major microinjecting of 5-aza-2-deoxycytidine into ventrolateral orbital
depressive disorder predicted by DNA methylation. cortex on depressive-like behavior in rats. Neurosci Lett 574:11–
Neuropsychobiology 75(2):81–88. https://doi.org/10.1159/ 14. https://doi.org/10.1016/j.neulet.2014.04.050
000480512 106. Martinowich K, Hattori D, Wu H, Fouse S, He F, Hu Y, Fan G,
91. Dwivedi Y, Rizavi HS, Conley RR, Roberts RC, Tamminga CA, Sun YE (2003) DNA methylation-related chromatin remodeling
Pandey GN (2003) Altered gene expression of brain-derived neu- in activity-dependent BDNF gene regulation. Science 302(5646):
rotrophic factor and receptor tyrosine kinase B in postmortem 890–893. https://doi.org/10.1126/science.1090842
brain of suicide subjects. Arch Gen Psychiatry 60(8):804–815. 107. Fuchikami M, Morinobu S, Segawa M, Okamoto Y, Yamawaki S,
https://doi.org/10.1001/archpsyc.60.8.804 Ozaki N, Inoue T, Kusumi I et al (2011) DNA methylation profiles
92. Tsankova N, Renthal W, Kumar A, Nestler EJ (2007) Epigenetic of the brain-derived neurotrophic factor (BDNF) gene as a potent
regulation in psychiatric disorders. Nat Rev Neurosci 8(5):355– diagnostic biomarker in major depression. PLoS One 6(8):e23881.
367. https://doi.org/10.1038/nrn2132 https://doi.org/10.1371/journal.pone.0023881
93. Krishnan V, Nestler EJ (2008) The molecular neurobiology of 108. Tsankova NM, Berton O, Renthal W, Kumar A, Neve RL, Nestler
depression. Nature 455(7215):894–902. https://doi.org/10.1038/ EJ (2006) Sustained hippocampal chromatin regulation in a mouse
nature07455 model of depression and antidepressant action. Nat Neurosci 9(4):
94. Zhu K, Ou Yang TH, Dorie V, Zheng T, Anastassiou D (2019) 519–525. https://doi.org/10.1038/nn1659
Meta-analysis of expression and methylation signatures indicates 109. Kang HJ, Kim JM, Lee JY, Kim SY, Bae KY, Kim SW, Shin IS,
a stress-related epigenetic mechanism in multiple neuropsychiatric Kim HR et al (2013) BDNF promoter methylation and suicidal
disorders. Transl Psychiatry 9(1):32. https://doi.org/10.1038/ behavior in depressive patients. J Affect Disord 151(2):679–685.
s41398-018-0358-5 https://doi.org/10.1016/j.jad.2013.08.001
95. Jones PA, Takai D (2001) The role of DNA methylation in mam- 110. Chan RF, Turecki G, Shabalin AA, Guintivano J, Zhao M, Xie
malian epigenetics. Science 293(5532):1068–1070. https://doi. LY, van Grootheest G, Kaminsky ZA et al (2020) Cell type-
org/10.1126/science.1063852 specific methylome-wide association studies implicate
96. Tognini P, Napoli D, Pizzorusso T (2015) Dynamic DNA meth- neurotrophin and innate immune signaling in major depressive
ylation in the brain: a new epigenetic mark for experience- disorder. Biol Psychiatry 87(5):431–442. https://doi.org/10.1016/
dependent plasticity. Front Cell Neurosci 9:331. https://doi.org/ j.biopsych.2019.10.014
10.3389/fncel.2015.00331 111. Chang LC, Jamain S, Lin CW, Rujescu D, Tseng GC, Sibille E
97. Wang P, Lv Q, Mao Y, Zhang C, Bao C, Sun H, Chen H, Yi Z et al (2014) A conserved BDNF, glutamate- and GABA-enriched gene
(2018) HTR1A/1B DNA methylation may predict escitalopram module related to human depression identified by coexpression
treatment response in depressed Chinese Han patients. J Affect meta-analysis and DNA variant genome-wide association studies.
Disord 228:222–228. https://doi.org/10.1016/j.jad.2017.12.010 PLoS One 9(3):e90980. https://doi.org/10.1371/journal.pone.
98. Le Francois B, Soo J, Millar AM, Daigle M, Le Guisquet AM, 0090980
Leman S, Minier F, Belzung C et al (2015) Chronic mild stress 112. Keller S, Sarchiapone M, Zarrilli F, Tomaiuolo R, Carli V,
and antidepressant treatment alter 5-HT1A receptor expression by Angrisano T, Videtic A, Amato F et al (2011) TrkB gene expres-
modifying DNA methylation of a conserved Sp4 site. Neurobiol sion and DNA methylation state in Wernicke area does not asso-
Dis 82:332–341. https://doi.org/10.1016/j.nbd.2015.07.002 ciate with suicidal behavior. J Affect Disord 135(1–3):400–404.
99. Gassen NC, Fries GR, Zannas AS, Hartmann J, Zschocke J, https://doi.org/10.1016/j.jad.2011.07.003
Hafner K, Carrillo-Roa T, Steinbacher J et al (2015) 113. Wang P, Zhang C, Lv Q, Bao C, Sun H, Ma G, Fang Y, Yi Z et al
Chaperoning epigenetics: FKBP51 decreases the activity of (2018) Association of DNA methylation in BDNF with
DNMT1 and mediates epigenetic effects of the antidepressant escitalopram treatment response in depressed Chinese Han pa-
paroxetine. Sci Signal 8(404):ra119. https://doi.org/10.1126/ tients. Eur J Clin Pharmacol 74(8):1011–1020. https://doi.org/10.
scisignal.aac7695 1007/s00228-018-2463-z
100. Higuchi F, Uchida S, Yamagata H, Otsuki K, Hobara T, Abe N, 114. Aberg KA, Dean B, Shabalin AA, Chan RF, Han LKM, Zhao M,
Shibata T, Watanabe Y (2011) State-dependent changes in the van Grootheest G, Xie LY et al (2018) Methylome-wide associa-
expression of DNA methyltransferases in mood disorder patients. tion findings for major depressive disorder overlap in blood and
J Psychiatr Res 45(10):1295–1300. https://doi.org/10.1016/j. brain and replicate in independent brain samples. Mol Psychiatry.
jpsychires.2011.04.008 https://doi.org/10.1038/s41380-018-0247-6
101. Sales AJ, Biojone C, Terceti MS, Guimaraes FS, Gomes MV, Joca 115. Boersma GJ, Lee RS, Cordner ZA, Ewald ER, Purcell RH,
SR (2011) Antidepressant-like effect induced by systemic and Moghadam AA, Tamashiro KL (2014) Prenatal stress decreases
intra-hippocampal administration of DNA methylation inhibitors. Bdnf expression and increases methylation of Bdnf exon IV in
Br J Pharmacol 164(6):1711–1721. https://doi.org/10.1111/j. rats. Epigenetics 9(3):437–447. https://doi.org/10.4161/epi.27558
1476-5381.2011.01489.x 116. Chandrasekar R (2013) Alcohol and NMDA receptor: current re-
102. Sales AJ, Joca SR (2016) Effects of DNA methylation inhibitors search and future direction. Front Mol Neurosci 6:14. https://doi.
and conventional antidepressants on mice behaviour and brain org/10.3389/fnmol.2013.00014
Mol Neurobiol

117. Kaut O, Schmitt I, Hofmann A, Hoffmann P, Schlaepfer TE, 134. Feng J, Zhou Y, Campbell SL, Le T, Li E, Sweatt JD, Silva AJ,
Wullner U, Hurlemann R (2015) Aberrant NMDA receptor Fan G (2010) Dnmt1 and Dnmt3a maintain DNA methylation and
DNA methylation detected by epigenome-wide analysis of hippo- regulate synaptic function in adult forebrain neurons. Nat
campus and prefrontal cortex in major depression. Eur Arch Neurosci 13(4):423–430. https://doi.org/10.1038/nn.2514
Psychiatry Clin Neurosci 265(4):331–341. https://doi.org/10. 135. Levenson JM, Roth TL, Lubin FD, Miller CA, Huang IC, Desai P,
1007/s00406-014-0572-y Malone LM, Sweatt JD (2006) Evidence that DNA (cytosine-5)
118. Jobe EM, Zhao X (2017) DNA methylation and adult methyltransferase regulates synaptic plasticity in the hippocam-
neurogenesis. Brain Plast 3(1):5–26. https://doi.org/10.3233/ pus. J Biol Chem 281(23):15763–15773. https://doi.org/10.1074/
BPL-160034 jbc.M511767200
119. Sweatt JD (2016) Dynamic DNA methylation controls glutamate 136. Poulter MO, Du L, Weaver IC, Palkovits M, Faludi G, Merali Z,
receptor trafficking and synaptic scaling. J Neurochem 137(3): Szyf M, Anisman H (2008) GABAA receptor promoter hyperme-
312–330. https://doi.org/10.1111/jnc.13564 thylation in suicide brain: Implications for the involvement of
120. Mahar I, Bambico FR, Mechawar N, Nobrega JN (2014) Stress, epigenetic processes. Biol Psychiatry 64(8):645–652. https://doi.
serotonin, and hippocampal neurogenesis in relation to depression org/10.1016/j.biopsych.2008.05.028
and antidepressant effects. Neurosci Biobehav Rev 38:173–192. 137. Hsieh MT, Lin CC, Lee CT, Huang TL (2019) Abnormal brain-
https://doi.org/10.1016/j.neubiorev.2013.11.009 derived neurotrophic factor exon IX promoter methylation, pro-
121. Fukumoto K, Iijima M, Funakoshi T, Chaki S (2018) Role of 5- tein, and mRNA levels in patients with major depressive disorder.
HT1A receptor stimulation in the medial prefrontal cortex in the J Clin Med 8(5). https://doi.org/10.3390/jcm8050568
sust aine d ant ide pres sant effects of ket amine. Int J 138. Efstathopoulos P, Andersson F, Melas PA, Yang LL, Villaescusa
Neuropsychopharmacol 21(4):371–381. https://doi.org/10.1093/ JC, Ruegg J, Ekstrom TJ, Forsell Y et al (2018) NR3C1 hyperme-
ijnp/pyx116 thylation in depressed and bullied adolescents. Transl Psychiatry
122. Zhou W, Wang N, Yang C, Li XM, Zhou ZQ, Yang JJ (2014) 8(1):121. https://doi.org/10.1038/s41398-018-0169-8
Ketamine-induced antidepressant effects are associated with 139. Haghighi F, Xin Y, Chanrion B, O’Donnell AH, Ge Y, Dwork AJ,
AMPA receptors-mediated upregulation of mTOR and BDNF in Arango V, Mann JJ (2014) Increased DNA methylation in the
rat hippocampus and prefrontal cortex. Eur Psychiatry 29(7):419– suicide brain. Dialogues Clin Neurosci 16(3):430–438
423. https://doi.org/10.1016/j.eurpsy.2013.10.005 140. Morris MJ, Na ES, Autry AE, Monteggia LM (2016) Impact of
123. Sales A, Biojone C, Joca S (2016) Site-specific delivery of epige- DNMT1 and DNMT3a forebrain knockout on depressive- and
netic modulating drugs into the rat brain. In: Karpova N (ed) anxiety like behavior in mice. Neurobiol Learn Mem 135:139–
Epigenetic methods in neuroscience research, Neuromethods, 145. https://doi.org/10.1016/j.nlm.2016.08.012
vol 105. Humana press, New York 141. Elliott E, Manashirov S, Zwang R, Gil S, Tsoory M, Shemesh Y,
124. Paxinos G, Watson C (2013) The rat brain in stereotaxic coordi- Chen A (2016) Dnmt3a in the medial prefrontal cortex regulates
nates, 7th edn. Academic Press, Cambridge anxiety-like behavior in adult mice. J Neurosci 36(3):730–740.
125. Abel EL, Bilitzke PJ (1990) A possible alarm substance in the https://doi.org/10.1523/JNEUROSCI.0971-15.2016
forced swimming test. Physiol Behav 48(2):233–239 142. Momparler RL (2005) Pharmacology of 5-Aza-2′-deoxycytidine
126. Bonefeld BE, Elfving B, Wegener G (2008) Reference genes for (decitabine). Semin Hematol 42(3 Suppl 2):S9–S16
normalization: a study of rat brain tissue. Synapse 62(4):302–309. 143. Fandy TE (2009) Development of DNA methyltransferase inhib-
https://doi.org/10.1002/syn.20496 itors for the treatment of neoplastic diseases. Curr Med Chem
127. Karpova NN, Umemori J (2016) Protocol for methylated DNA 16(17):2075–2085. https://doi.org/10.2174/
immunoprecipitation (meDIP) analysis. In: Epigenetic methods in 092986709788612738
neuroscience research, vol 105. Humana Press, New York, pp. 144. Brueckner B, Garcia Boy R, Siedlecki P, Musch T, Kliem HC,
97–114 Zielenkiewicz P, Suhai S, Wiessler M et al (2005) Epigenetic
128. Roy B, Shelton RC, Dwivedi Y (2017) DNA methylation and reactivation of tumor suppressor genes by a novel small-
expression of stress related genes in PBMC of MDD patients with molecule inhibitor of human DNA methyltransferases. Cancer
and without serious suicidal ideation. J Psychiatr Res 89:115–124. Res 65(14):6305–6311. https://doi.org/10.1158/0008-5472.
https://doi.org/10.1016/j.jpsychires.2017.02.005 CAN-04-2957
129. Casarotto PC, de Bortoli VC, Correa FM, Resstel LB, Zangrossi H 145. Schirrmacher E, Beck C, Brueckner B, Schmitges F, Siedlecki P,
Jr (2010) Panicolytic-like effect of BDNF in the rat dorsal Bartenstein P, Lyko F, Schirrmacher R (2006) Synthesis and
periaqueductal grey matter: the role of 5-HT and GABA. Int J in vitro evaluation of biotinylated RG108: a high affinity com-
Neuropsychopharmacol 13(5):573–582. https://doi.org/10.1017/ pound for studying binding interactions with human DNA meth-
S146114570999112X yltransferases. Bioconjug Chem 17(2):261–266. https://doi.org/
130. Fernandez Macedo GV, Cladouchos ML, Sifonios L, Cassanelli 10.1021/bc050300b
PM, Wikinski S (2013) Effects of fluoxetine on CRF and CRF1 146. Yang C, Shirayama Y, Zhang JC, Ren Q, Yao W, Ma M, Dong C,
expression in rats exposed to the learned helplessness paradigm. Hashimoto K (2015) R-ketamine: a rapid-onset and sustained an-
Psychopharmacology 225(3):647–659. https://doi.org/10.1007/ tidepressant without psychotomimetic side effects. Transl
s00213-012-2859-x Psychiatry 5:e632. https://doi.org/10.1038/tp.2015.136
131. Yan HC, Cao X, Das M, Zhu XH, Gao TM (2010) Behavioral 147. Schneeberger Y, Stenzig J, Hubner F, Schaefer A, Reichenspurner
animal models of depression. Neurosci Bull 26(4):327–337. H, Eschenhagen T (2016) Pharmacokinetics of the experimental
https://doi.org/10.1007/s12264-010-0323-7 non-nucleosidic DNA methyl transferase inhibitor N-phthalyl-L-
132. Urb M, Anier K, Matsalu T, Aonurm-Helm A, Tasa G, Koppel I, tryptophan (RG 108) in rats. Basic Clin Pharmacol Toxicol
Zharkovsky A, Timmusk T et al (2019) Glucocorticoid receptor 118(5):327–332. https://doi.org/10.1111/bcpt.12514
stimulation resulting from early life stress affects expression of 148. Marcucci G, Silverman L, Eller M, Lintz L, Beach CL (2005)
DNA methyltransferases in rat prefrontal cortex. J Mol Neurosci Bioavailability of azacitidine subcutaneous versus intravenous in
68(1):99–110. https://doi.org/10.1007/s12031-019-01286-z patients with the myelodysplastic syndromes. J Clin Pharmacol
133. Morris MJ, Adachi M, Na ES, Monteggia LM (2014) Selective 45(5):597–602. https://doi.org/10.1177/0091270004271947
role for DNMT3a in learning and memory. Neurobiol Learn Mem 149. Champagne FA, Francis DD, Mar A, Meaney MJ (2003)
115:30–37. https://doi.org/10.1016/j.nlm.2014.06.005 Variations in maternal care in the rat as a mediating influence
Mol Neurobiol

for the effects of environment on development. Physiol Behav 160. Roth TL, Lubin FD, Funk AJ, Sweatt JD (2009) Lasting epige-
79(3):359–371 netic influence of early-life adversity on the BDNF gene. Biol
150. Li N, Liu RJ, Dwyer JM, Banasr M, Lee B, Son H, Li XY, Psychiatry 65(9):760–769. https://doi.org/10.1016/j.biopsych.
Aghajanian G et al (2011) Glutamate N-methyl-D-aspartate recep- 2008.11.028
tor antagonists rapidly reverse behavioral and synaptic deficits 161. Zhang JC, Yao W, Dong C, Yang C, Ren Q, Ma M, Han M,
caused by chronic stress exposure. Biol Psychiatry 69(8):754– Hashimoto K (2015) Comparison of ketamine, 7,8-
761. https://doi.org/10.1016/j.biopsych.2010.12.015 dihydroxyflavone, and ANA-12 antidepressant effects in the so-
151. Ardalan M, Rafati AH, Nyengaard JR, Wegener G (2017) Rapid cial defeat stress model of depression. Psychopharmacology
antidepressant effect of ketamine correlates with astroglial plastic- 232(23):4325–4335. https://doi.org/10.1007/s00213-015-4062-3
ity in the hippocampus. Br J Pharmacol 174(6):483–492. https:// 162. Shirayama Y, Hashimoto K (2018) Lack of antidepressant effects
doi.org/10.1111/bph.13714 of (2R,6R)-hydroxynorketamine in a rat learned helplessness
152. Ardalan M, Wegener G, Rafati AH, Nyengaard JR (2017) S- model: comparison with (R)-ketamine. Int J
ketamine rapidly reverses synaptic and vascular deficits of hippo- Neuropsychopharmacol 21(1):84–88. https://doi.org/10.1093/
campus in genetic animal model of depression. Int J ijnp/pyx108
Neuropsychopharmacol 20(3):247–256. https://doi.org/10.1093/ 163. Maussion G, Yang J, Suderman M, Diallo A, Nagy C, Arnovitz
ijnp/pyw098 M, Mechawar N, Turecki G (2014) Functional DNA methylation
153. Ardalan M, Elfving B, Rafati AH, Mansouri M, Zarate CA Jr, in a transcript specific 3′UTR region of TrkB associates with sui-
Mathe AA, Wegener G (2020) Rapid effects of S-ketamine on cide. Epigenetics 9(8):1061–1070. https://doi.org/10.4161/epi.
the morphology of hippocampal astrocytes and BDNF serum 29068
levels in a sex-dependent manner. Eur Neuropsychopharmacol 164. Mifsud KR, Saunderson EA, Spiers H, Carter SD, Trollope AF,
32:94–103. https://doi.org/10.1016/j.euroneuro.2020.01.001 Mill J, Reul JM (2017) Rapid down-regulation of glucocorticoid
154. Lepack AE, Fuchikami M, Dwyer JM, Banasr M, Duman RS receptor gene expression in the dentate gyrus after acute stress
(2014) BDNF release is required for the behavioral actions of in vivo: role of DNA methylation and microRNA activity.
ketamine. Int J Neuropsychopharmacol 18(1). https://doi.org/10. Neuroendocrinology 104(2):157–169. https://doi.org/10.1159/
1093/ijnp/pyu033 000445875
155. Lener MS, Kadriu B, Zarate CA Jr (2017) Ketamine and beyond:
165. Martin-Hernandez D, Tendilla-Beltran H, Madrigal JLM, Garcia-
investigations into the potential of glutamatergic agents to treat
Bueno B, Leza JC, Caso JR (2019) Chronic mild stress alters
depression. Drugs 77(4):381–401. https://doi.org/10.1007/
kynurenine pathways changing the glutamate neurotransmission
s40265-017-0702-8
in frontal cortex of rats. Mol Neurobiol 56(1):490–501. https://doi.
156. Hashimoto K (2011) The role of glutamate on the action of anti-
org/10.1007/s12035-018-1096-7
depressants. Prog Neuro-Psychopharmacol Biol Psychiatry 35(7):
166. van Eijk KR, de Jong S, Boks MP, Langeveld T, Colas F, Veldink
1558–1568. https://doi.org/10.1016/j.pnpbp.2010.06.013
JH, de Kovel CG, Janson E et al (2012) Genetic analysis of DNA
157. Castren E (2005) Is mood chemistry? Nat Rev Neurosci 6(3):241–
methylation and gene expression levels in whole blood of healthy
246. https://doi.org/10.1038/nrn1629
human subjects. BMC Genomics 13:636. https://doi.org/10.1186/
158. Courtney MJ, Akerman KE, Coffey ET (1997) Neurotrophins
1471-2164-13-636
protect cultured cerebellar granule neurons against the early phase
of cell death by a two-component mechanism. J Neurosci 17(11): 167. Lea AJ, Vockley CM, Johnston RA, Del Carpio CA, Barreiro LB,
4201–4211 Reddy TE, Tung J (2018) Genome-wide quantification of the
159. Karege F, Vaudan G, Schwald M, Perroud N, La Harpe R (2005) effects of DNA methylation on human gene regulation. eLife 7.
Neurotrophin levels in postmortem brains of suicide victims and https://doi.org/10.7554/eLife.37513
the effects of antemortem diagnosis and psychotropic drugs. Brain
Res Mol Brain Res 136(1–2):29–37. https://doi.org/10.1016/j. Publisher’s Note Springer Nature remains neutral with regard to jurisdic-
molbrainres.2004.12.020 tional claims in published maps and institutional affiliations.

You might also like