You are on page 1of 17

Review

For reprint orders, please contact: reprints@futuremedicine.com


Pharmacogenomics

Pharmacogenetics in electroconvulsive
therapy and adjunctive medications

Electroconvulsive therapy (ECT) has shown apparent efficacy in treatment of patients Hooman Mirzakhani1,2,3,
with depression and other mental illnesses who do not respond to psychotropic Martijn S van Noorden4,
medications or need urgent control of their symptoms. Pharmacogenetics contributes Jesse Swen3, Ala Nozari5,6
to an individual’s sensitivity and response to a variety of drugs. Clinical insights into & Henk-Jan Guchelaar*,3
1
Channing Division of Network Medicine,
pharmacogenetics of ECT and adjunctive medications not only improves its safety Department of Medicine, Brigham &
and efficacy in the indicated patients, but can also lead to the identification of novel Women’s Hospital, Boston, MA, USA
treatments in psychiatric disorders through understanding of potential molecular 2
Division of Biomedical Informatics,
and biological mechanisms involved. In this review, we explore the indications of Harvard Medical School, Boston, MA,
USA
pharmacogenetics role in safety and efficacy of ECT and present the evidence for its 3
Department of Clinical Pharmacy &
role in patients with psychiatric disorders undergoing ECT. Toxicology, Leiden University Medical
Center, Leiden University, Leiden,
Keywords: depression • ECT • efficacy • pharmacogenetics • safety The Netherlands
4
Department of Psychiatry, Leiden
University Medical Center, Leiden
Due to its apparent effectiveness, electro­ ment procedures via multiple components. University, Leiden, The Netherlands
convulsive therapy (ECT) was commonly This interface between our genetic variations 5
Department of Anesthesia, Orthopedic
used for depressive disorders prior to inven­ and response to therapeutic interventions, in Anesthesia Division, Massachusetts
tion of antidepressants in the 1950s. In the other words, pharmacogenetics, determines General Hospital, Boston, MA, USA
6
Harvard Medical School, Boston, MA,
beginning, unmodified ECT was agonizing the individual response to a specific treat­
USA
and unsafe, leading to, among others, bone ment and affects both patient’s safety and *Author for correspondence:
fracture or spine injury. However, the advent efficacy of response to ECT. Tel.: +31 071 526 2790
of anesthetics and muscle relaxants along Historically, the role of pharmaco­genetics h.j.guchelaar@ lumc.nl
with newer apparatus evolved the modified in ECT was primarily related to the safety
ECT to achieve maximum benefits while of ECT application and was explored by
minimizing the procedural side effects related Kalow. Observation of prolonged apnea after
to the applied medication (anesthesia), elec­ succinylcholine administration in some indi­
trical current or the induced seizure. ECT viduals paved the way to the preliminary
is currently administered with individually clinical insights into pharmacogenetics. In
adjusted electrical currents under the surveil­ 1956, Kalow discovered that an alteration
lance of anesthesiologists who apply anesthet­ in plasma cholinesterase level caused induc­
ics and muscle relaxants in an appropriate tion of longer duration of paralysis by appli­
medical setting [1] . cation of succinylcholine to ECT patients.
In spite of all the technical advances that This change resulted in extension of dura­
have improved the relative risk of ECT, tion of succinylcholine action from the few
many crucial issues remain unresolved. For minutes to over an hour in the affected indi­
example, it is difficult to determine a priori viduals [3,4] . Kalow implied the discovery
whether ECT will be associated with an of the presence of genetic effects on drug
‘adequate’ therapeutically effective seizure in response [5,6] .
each patient [2] . Genetic variations contribute These studies advanced further research
to the variation of an individual’s response related to pharmacogenetics such that in 1960s
part of
to a different class of medications or treat­ the process of drug acetylation was discovered

10.2217/PGS.15.57 © 2015 Future Medicine Ltd Pharmacogenomics (Epub ahead of print) ISSN 1462-2416
Review Mirzakhani, Noorden, Swen, Nozari & Guchelaar

and the fact that slow acetylators (poor metabolizers) are words ‘ECT anesthesia genetics’, ‘ECT safety genetics’,
more prone to side effects related to drug metabolism [5] . ‘ECT efficacy genetics’, ‘ECT and genetics’, ‘treatment-
Furthermore, by application of anesthetics during pro­ resistant psychotic disorders ECT genetics’, ‘depressive
cedural treatments including ECT, the awareness of disorders ECT genetics’, ‘psychotic disorders ECT
developing side effects such as acute porphyric event due genetics’, ‘ECT neurotransmitter genes’, ‘genetics and
to thiopentone as well as the observed association of the ECT response’ and ‘adjunctive therapy in ECT and
malignant hyperthermia with succinylcholine, drew the genetics’. References of the relevant articles or edito­
attention in how genetics might influence in the applied rials were also considered for potential bibliographic-
pharmacology [7] . related references to avoid any missing publication. All
ECT is the most effective acute treatment for mood searches were limited to research published in English.
disorders, especially treatment-resistant major depres­ Due to paucity of articles on this subject, there was
sive disorder (MDD), and has proven efficacy in other no restriction on time of publication. The identified
nondepressed psychiatric disorders such as schizophre­ papers were predominantly related to depression dis­
nia [8–11] . Several studies have explored the impact of orders. For the efficacy of ECT and main focus of this
genetics in the neurobiological mechanisms involved review, we included all preclinical and human studies
in ECT. These studies have also aimed to improve the that might explain a direct role of a gene in the efficacy
efficacy of ECT by predicting the genotype-phenotype of ECT or suggest a genetic effect through neurobio­
distinction of patients undergoing ECT [2,12,13] . To our logical mechanisms. Accordingly, 34 publications were
knowledge, no comprehensive literature review has identified in investigating the pharmacogenetics in
been published on this ground. Hence, in this review, efficacy of ECT; only 19 articles showed direct investi­
we will provide an evidence-based approach to the gation on role of pharmacogentics in ECT. The ECT-
role of pharmacogenetics on safety (drugs used during associated gene signatures were analyzed using Meta­
ECT) and efficacy of ECT (potential genes and neu­ Core™ platform for investigation of their potential
robiological mechanisms) and present evidence of its interactions and common biological network pathways
importance in patients with psychiatric disorders who in human brain.
undergo ECT (Figure 1) .
Pharmacogenetics & safety of ECT
Methods Anesthesia adverse events
Using a structured approach to identify the source of Abnormalities in butyrylcholinesterase
materials for the review, a systematic search was con­ ECT is a short procedure and demands anesthetics with
ducted for relevant peer-reviewed articles in PubMed rapid onset and short duration of action. These qualities
and the Google Scholar search engine, using the key­ have made succinylcholine as the neuromuscular block­

Potential role of pharmacogenetics in patients indicated for ECT

Safety of procedure Efficacy of procedure

Peri-ECT Post-ECT

Anesthesia adverse events


NMDA receptor activation and Chronic molecular effects of ECT
induced pathway, cortisol (BDNF, VEGF, TRKB, CREB-
activated genes, DARPP32,
CACNA2D1 and CACNA1S, BCHE Acute molecular effects of ECT 5-HT2 receptors, AP-1 complex)
genotype, CYP2D6 and RYR1 (glutamate, aspartate, GABA,
tryptophan production pathways)

Prediction of the genotype–


phenotype distinction of patients
undergoing ECT (DDR2, COMT,
DRD2, DRD3)

Figure 1. Various implications of pharmacogenetics in safety and efficacy of electroconvulsive therapy.


ECT: Electroconvulsive therapy.

10.2217/PGS.15.57 Pharmacogenomics (Epub ahead of print) future science group


Pharmacogenetics in electroconvulsive therapy & adjunctive medications Review

ing agent of choice for ECT [1] . The enzyme butyryl­ duration of apnea were prolonged (5–15 min) in 11
cholinesterase (BCHE) hydrolyzes ester bonds in suc­ patients compared with controls (3–5.3 min). Conse­
cinylcholine. The enzyme is composed of four identical quently, they recommended neuromuscular monitoring
subunits of 574 amino acids, each containing an active during the first ECT.
catalytic site. The BCHE1 gene is located on four
exons of chromosome 3q26.1–3q26.2 (online Mende­ Malignant hyperthermia
lian inheritance in Man; OMIM 177,400). The gene Malignant hyperthermia (MH) is a hypermetabolic
encodes a 602 amino acid protein including a 28 amino response to succinylcholine and potent volatile anes­
acid leader peptide. Genetic variation in the BCHE1 thetic gases such as halothane, isoflurane, sevoflurane
gene leads to variant enzyme forms, which affect the and desflurane. Succinylcholine, the main applied
substrate behavior, resulting in reduced or absence of muscle relaxant in ECT, is a far more powerful trigger
the enzyme BCHE activity. BCHE deficiency results in of MH than the volatile agents [22–24] . Experimental
a prolonged effect of the ultra-short acting depolarizing studies indicate that the mechanism underlying MH
muscle relaxant succinylcholine due to marked decrease is an uncontrolled release of intracellular calcium from
in plasma cholinesterase activity [14] . The variants of skeletal muscle sarcoplasmic reticulum (SR). MH is
BCHE included wild-type (U), atypical (A, dibucaine inherited primarily in an autosomal-dominant fashion
resistant), fluoride resistant (F), silent (S) and Kalow in humans, which might result in the MH prevalence
(K). Besides the normal variant (U), A and K variants of up to 1:3000 due to the causal genetic mutation.
are more frequent (A20G and G1615A) [15] . It is noted A more complex inheritance pattern might also be
that the patients with homo­zygosis or carriers with observed in the affected individuals [25] .
compound heterozygosity remain asymptomatic in the Approximately 50% of MH-related genetic variants
absence of exposure ­exogenous ­choline ester-compound have been found in the RYR1 gene on chromosome 19,
such as succinylcholine. a calcium channel located in the SR. Most cases (70%)
In Australian population, out of 65 patients referred carry one of 30 RYR1 mutations. Linkage studies have
for prolonged postsuccinlycholine apnea, 85% of the implicated five other regions, with variants identified in
subjects showed one of the mutated variants of BCHE calcium channels CACNA2D1 and CACNA1S. While
gene including dibucaine (Dib; D70G), Sil-1 (G115FS), these regions account for genetic variants in less than 2%
Flu-1 (T243M), Flu-2 (G390V) and K-variant (A539T), of cases, the causal genetic variant in approximately 30%
with 74% being dibucaine homozygote or heterozygote, of patients remains unknown [26,27] . Genetic tests may
6% rare genotypes, 3% heterozygous fluoride allele and offer a noninvasive diagnostic method with lower mor­
13% undetermined [16] . The Danish Cholinesterase bidity than in vitro muscle contracture tests as the cur­
Research Unit (DCRU), in a 20-year longitudinal study, rent functional gold standard for MH diagnosis; how­
found abnormal response to succinylcholine in 61.1% of ever, genetic testing is unreliable because the spectrum of
the 1247 patients who were visited in the center. Out of contributing loci and alleles is not yet fully understood.
these 1247 patients, 28.5% had normal genotype and The reported incidence of MH during ECT has
46.5% were genotypically aberrant. While the recovery been less than other procedures requiring general anes­
time of neuromuscular function in patients with one thesia. It seems more likely that other factors may be
aberrant allele was 15–30 min following a single dose of responsible for this observation. Gornert indicated that
succinylcholine 1.0–1.5 mg kg-1, it took 35–45 min for MH is triggered in proportion to the total dose of trig­
patients with heterozygosity of abnormal allele in two gering agents [28] . Thus, even if MH is triggered by
genes to recover. Homozygosity in abnormal allele in succinylcholine during ECT, the absence of continued
atypical gene lengthened the recovery time to 90–180 administration of succinylcholine may abort its more
min. Patients with genotypes of AK and AH experi­ fulminant expression. Indeed, the anesthetics used
enced slightly (20 min) or markedly longer duration in ECT are almost always ultra-short or short-acting
of action (90 min) of succinylcholine, respectively [17] . barbiturates such as methohexital or propofol. Absence
Accordingly, there are several case reports of succinyl­ of the use of volatile anesthetics in ECT also may
choline-induced prolonged apnea in patients receiving explain the lack of induction of MH. As was previously
ECT [1,18–20] . In a longitudinal study, Mollerup et al. stated, potent volatile anesthetics that are commonly
determined the BCHE activity and the BCHE geno­ employed in general anesthesia are also strongly linked
type of 13 patients who were visited in the DCRU after to precipitation of MH [29] .
ECT [21] . The authors measured and compared the dura­
tion of apnea with normal subjects and found BCHE Neuroleptic malignant syndrome
gene mutations, the K-variant being the most frequent. Neuroleptic malignant syndrome (NMS) is a serious
The duration of succinylcholine action and consequently and potentially fatal side effect of anti­psychotics, com­

future science group 10.2217/PGS.15.57


Review Mirzakhani, Noorden, Swen, Nozari & Guchelaar

prising fever, muscle rigidity, delirium and autonomic Pharmacogenetics & the efficacy of ECT
dysfunction. Underlying mechanism of this side effect Effect of adjunctive psychotropic drugs on ECT
is still unknown and debated. So far some risk factors outcome
have been identified, with clinical observations and The current remission rates after ECT appear to have
recent pharmacogenetic research suggesting, though declined [8,41] . In a meta-analysis to investigate the
with inconsistent findings, correlation between effect of previous pharmacotherapy failure on the effi­
genetic mechanisms and predisposition to NMS [30] . cacy of ECT, the overall remission rate was reported
Polymorphisms of CYP2D6 enzyme through which to be 48.0% (281 of 585) and 64.9% (242 of 373)
most psychotropic drugs are metabolized and TaqIA for patients with and without previous pharmaco­
DRD2, a target for antipsychotic drugs, has been therapy failure, respectively. Additionally, patients
reported to act as the link between pharmacogenetic who received previous pharmacotherapy but failed
factors and the potential development of NMS [30] . In to respond to the treatment, showed reduced efficacy
spite of these genetic links, ECT has been reported of ECT [42] . ECT has shown higher efficacy in con­
to be useful for refractory NMS or improving NMS junction with antidepressants and antipsychotics [43] .
symptoms [31,32] . Further investigation in an appro­ Accordingly, the understanding of pharmacogenetics
priately designed study is warranted to investigate the of the adjunctive drug therapy and more comprehen­
treatment role of ECT in NMS and identify the subset sively pharmacogenomics of such treatments has the
of patients with NMS who might benefit from this potential to improve therapeutic outcomes of ECT
procedure. and individualized drug therapy, while avoiding toxic
effects and treatment failure.
Potential neurobiological mechanisms in Genetic predictors of antipsychotics have been
cognitive side effects of ECT widely studied [44] , but fewer evidences are available
There are some animal and human preliminary evi­ how these factors might influence their role in the
dences on the role of neurotransmitters/biologi­ outcome of ECT.
cal alterations with the adverse cognitive effect of Some studies provide evidence that the applica­
ECT [33,34] . The investigated systematic alterations tion of the NMDA receptor antagonist ketamine
include cholinergic, endogenous opioid, glucocorti­ (0.5 mg/kg) could provide rapid and longer antide­
coid and glutaminergic systems, which were mostly pressant effects after ECT [45–50] . These studies have
conducted in animal models [34–37] . Among these, suggested the involvement of synaptic plasticity and
the effects of glutaminergic and glucocorticoid com­ neurotrophic signaling in the mechanism of action of
pounds have been investigated in human studies. In an ketamine. The observation of mammalian target of
ECT trial of ten subjects, using ketamine as anesthet­ rapamycin (mTOR) and brain-derived neurotrophic
ics resulted in less impairment of short-term memory factor (BDNF) pathway activation by NMDA recep­
than applying etomidate [37] . The result is suggestive tor antagonism has proposed the observed link for the
that the ECT-induced cognitive disruption might antidepressant action of ketamine through the interac­
be mediated by glutamate at N-methyl-D-aspartate tion between plasticity-related signaling pathway [51] .
receptors. Neylan et al. [38] , in a 2-week ECT trial of Using ketamine (0.5 mg/kg) has been promising in
16 subjects, showed that elevated basal level cortisol depressive patients prior to ECT [52,53] . Some evidences
was associated with a greater degree of ECT-induced that ketamine might reduce the effect of ECT on
cognitive impairment. memory has drawn more attention to the application
Palmio et al., in two separate studies, investigated of ketamine for ECT [37] . The optimal adjunctive dose
the acute effect of ECT on the perturbations in the range of ketamine, its safety and effective duration of
amino acid transmitters and the brain biomarkers action should be further investigated.
in blood with potential role in neuronal activity and The association between carrier status for the
neuronal injury. In treatment-resistant depression long allele of serotonin transporter gene with a bet­
(TRD)-MDD patients who underwent a single ECT ter response to serotonin selective reuptake inhibitors
session, they showed significant changes in the serum (SSRIs) has been of interest. Solute carrier family 6
levels of glutamate, aspartate, gamma-aminobutyric (neurotransmitter transporter, serotonin), member
acid (GABA), S-100b protein (S-100b), tryptophan 4 (SLC6A4) encodes the serotonin transporter and
and some other amino acids in 24 h [39,40] . However, is located on location 17q11.1-q12. Rasmussen et al.
it is important that future studies are designed to retrospectively studied whether the polymorphism of
better distinguish changes in the levels of these bio­ the serotonin transporter gene (5-HTT ) was associ­
markers related to both ECT side effects on brain and ated with differential treatment response in 83 ECT
therapeutic response. patients treated for depressive disorder [12] . No signifi­

10.2217/PGS.15.57 Pharmacogenomics (Epub ahead of print) future science group


Pharmacogenetics in electroconvulsive therapy & adjunctive medications Review

cant association was found between serotonin trans­ matter (WM) microstructures between the frontal and
porter gene allelic status with several characteristics of limbic areas [61–63] , such that WM abnormalities relate
ECT treatment, such as seizure length or threshold, to depression severity and TRD [64–67] . Accordingly,
number of treatments in a series and depression scale Lyden et al. demonstrated ECT effect on structural
ratings. plasticity within dorsal fronto-limbic pathways and
plasticity of WM relation to therapeutic response in
Potential pharmacogenomics (neurobiological depression [67] . It is unknown if there is any genetic
mechanisms) in the efficacy of ECT predisposition to these structural alteration, and future
Brain alterations in acute & chronic ECT response investigation is hence warranted.
As was previously stated, for many nonresponsive
patients to psychotropic drugs, ECT is an efficient Potential genes involved in ECT response & the
rapid intervention. However, the neurobiological related biological pathways
mechanisms for the efficacy of ECT remain unknown. In 1998, Fochtmann et al. showed that both hippo­
While some of the therapeutic response to ECT is campal A1-receptor, and cortical and striatal NMDA-
shortly observed after treatment, similar to psycho­ receptor bindings are associated with the quality of
tropic drugs, efficacy of ECT increases by repeating seizure (i.e., duration) [2] . Their study suggested that
treatment. This fact has made neurotransmitters and induced ECT-neurobiological mechanisms potentially
metabolic enzymes of greater interest for investiga­ related to some genes, might contribute to the desired
tion of ECT response. ECT affects wide range of therapeutic effect of ECT.
brain areas [54,55] , which the potential therapeutic While genetic pathway alterations by ECT and their
effects on these regions may be through structural, association with clinical parameters could provide
and/or biochemical changes. pivotal information, few studies have examined the
Several neurotransmitters have been investigated genetic approaches to neurobiology of ECT and the
for involvement in association with psychotic dis­ impact of pharmacogenomics on treatment response
orders. Yatham et al. [56] suggested the role of sero­ in ECT (Table 1) [68–74] . These studies have shown
tonin (5-hydroxytryptamine, 5-HT) dysfunction. that chronic molecular effects induced by ECT are
Using positron emission tomography (PET) study more likely to reveal the mechanisms of its therapeutic
in patients with bipolar disorders, they showed anti­ effects.
depressants downregulate 5-HT2 receptors in several
cortical regions. In a follow-up study in patients who Gene-expression signatures
treated with ECT due to refractory response to anti­ Studies have suggested that the therapeutic effects
depressants, they showed similar effect by ECT in of ECT might be due to mechanisms involving sev­
downregulation of brain 5-HT2 receptors in the lim­ eral amino acid transmitter changes in brain through
bic and prefrontal cortical brain areas [57] . Similarly in overexpression of their regulatory genes. Altar et al.
another study by Lanzenberger et al., PET scan showed investigated the effects of single versus repeated
substantial reduction of 5-HT1A receptor-binding electro­
convulsive seizure (ECS) exposure on gene
potential (BPND) almost across the entire cortex after transcription, in an animal model, to identify genes
one ECT, particularly in amygdala and anterior cingu­ and potential biochemical pathways that are associ­
late cortex [58] . However in another study, after several ated with the efficacy of chronic ECT [55] . Almost
ECT, BPND did not show consistent result with the 120 hippocampal and frontal genes were differen­
former study [59] . tially expressed within distinct pathways (particu­
Dopamine neurotransmitter has also been of interest larly BDNF-MAP kinase) in response to acute and
in understanding the refractoriness of response to treat­ chronic ECS. Of those, only 19 genes showed similar
ment in depressive disorder. Saijo et al. scanned seven expression in response to acute or chronic ECS. Brain-
MDD patients’ brain after six to seven ECTs using derived neurotrophic factor (BDNF), cyclooxygen­
PET to examine the effect of treatment on dopamine ase (COX)-2, neuronal activity-regulated pentraxin
D2 receptors. They found significant increase in D2 (Narp) and TGFβ-inducible early growth response
receptors of anterior cingulate of patients who received had co-directional changes in both brain regions.
ECT [60] . Although these studies show regulatory role They suggested that the genes that increase only with
of serotonergic and dopaminergic pathways in biologi­ chronic ECS are more likely to be associated with effi­
cal mechanisms involved in response to ECT, it is yet to cacy of ECT, including those of the BDNF-TRKB-
be discovered how ECT causes these alterations. MAP kinase pathway, arachidonic acid pathway,
A few studies on depressive disorders have explored VEGF, thyrotropin-releasing hormone (TRH), neu­
the hypothesis of altered connectivity within the white ropeptide Y (NPY) and regulators of ­neurogenesis [55] .

future science group 10.2217/PGS.15.57


Table 1. Pharmacogenetic studies related to the outcome of electroconvulsive therapy in animal and human subjects.
Author Journal/year Subjects Procedure Outcome measure Author’s conclusion Ref. Review
Nibuya et al. J. Neuroscience/ Male rats Single and ten Northern blot analysis of BDNF BDNF and TRKB mRNA [75]
1995 daily ECS and TRKB mRNA expression expression increased by chronic

10.2217/PGS.15.57
ECS in frontal cortex
Zetterstrom Brain Res. Mol. Brain Res./ Male rats Single and five Densitometric quantification Both acute and chronic [76]
et al. 1998 ECS over 10 days ECS increased BDNF mRNA
expression in brain, markedly
in the granule cell layer of the
dentate gyrus
Fochtmann J ECT/1998 Male rodents Suprathreshold Correlations between Quality of seizure could [2]
et al. ECS hippocampal A1-receptor be influenced by heritable
binding, cortical and striatal factors which might affect the
NMDA-receptor binding, and neurobiologic mechanisms
the modification of seizure
duration by caffeine
Newton et al. J. Neuroscience/2003 Male rats ECS treatment Microarray analysis on several Growth factor and angiogenic [77]

Pharmacogenomics (Epub ahead of print)


genes involved in growth factor signaling could have
and angiogenic-endothelial modulating role in response to
Mirzakhani, Noorden, Swen, Nozari & Guchelaar

signaling, including VEGF treatment by ECS


Altar et al. J. Neuroscience/2004 Rats ECS treatment Gene transcription induction Acute and chronic ECS induces [55]
measurement in the frontal differential expressions of
cortex and hippocampus several regulatory transcripts
(e.g., BDNF) in brain. Subset
of these transcripts shows
similarity by time and region
Rosa et al. Brain Res./2007 Rats Single ECS Measurement of DARPP-32 Chronic application of ECS [78]
treatment, eight expression in time series increases DARPP-32 levels in
ECS treatments striatum and hippocampus
Voleti et al. Biol. Psychiatry/2012 Male rats Ten ECS Microarray analysis to identify Chronic ECS causes CREB- [79]
treatments CREB promoters that are dependent increase of Fz6
influenced by chronic ECS mRNA levels
Segawa et al. Int. J. Rats Single and Measurement of hippocampal Induction of BDNF expression [80]
Neuropsychopharmacol./ 10-day levels of pro-BDNF, PC1 and t-PA is involved in therapeutic
2013 administration response to ECS
of ECS
Taliaz et al. Biol. Psychiatry/2013 Male rats ECT for 10 days BDNF measurement by enzyme- ECT significantly reduced [81]
linked immunosorbent assay ventral tegmental area
in knockdown rats for BDNF BDNF expression causing
in hippocampus or ventral antidepressant-like effects
tegmental area after ECT

future science group


ECS: Electroconvulsive seizure; ECT: Electroconvulsive therapy; HAM-D: Hamilton depression rating scale; MADRS: Montgomery and Åsberg Depression Rating Scale; MDD: Major depressive disorder; OR: Odds
ratio; TRD: Treatment-resistant depression.
Table 1. Pharmacogenetic studies related to the outcome of electroconvulsive therapy in animal and human subjects (cont.).
Author Journal/year Subjects Procedure Outcome measure Author’s conclusion Ref.
Bocchio- Eur. Patients with TRD- ECT treatments Serum BDNF levels at ECT time Serum BDNF levels increases by [74]
Chiavetto Neuropsychopharmacol./ MDD with serial ECTs (three-times a (T0), after ECT (T1) and 1 month chronic application of ECT
et al. 2006 week) after end of ECT (T2)

future science group


Huuhka et al. Neurosci. Lett./2008 118 (female/male: ECT treatments MADRS <8 as responders, TT genotype of DRD2 C957T [13]
64/54) white patients (three-times a subjects were genotyping by polymorphism has synergistic
with treatment- week) using Taqman® SNP Genotyping effect with COMT gene
resistant MDD treated Assay for DRD2 and COMT polymorphism Met/Met
with ECT vs 383 polymorphism genotype and is was associated
healthy controls with less remission rate after
ECT
Huuhka et al. J. ECT/2005 and Neurosci. 118 (female/male: ECT treatments MADRS <8 as responders, RGS4 APOE and RGS4 genotype [69,71]
Lett./2008 65/ 54) white MDD (three-times a and APOE genotyping was are not associated with ECT
patients week) performed by using fluorogenic response in depression
allele-specific TaqMan probes
and primers
Anttila et al. Pharmacogenomics 119 treatment- ECT treatments MADRS <8 as responders, COMT high/high genotype [72]
J./2008 resistant MDD (three-times a patients’ DNA samples carriers are more likely to
patients (female/male: week) were genotyped by the respond to ECT (OR: 4.366 (95%
65/54) 5′ exonuclease assay CI: 1.137–16.770; p = 0.023)
Stewart et al. Neurosci. Lett./2009 119 treatment- ECT treatments Change in MADRS as the ACE genotype is not associated [82]
resistant MDD with (three-times a measure of treatment efficacy, with ECT outcome in depression
392 controls week) to compare the effects of the but might be associated with
ACE genotype distributions and age of onset
treatment response to ECT
Domschke Am. J. Med. Genet. Part B, 104 white TRD-MDD ECT treatments HAM-D change >50% baseline COMT gene variation might [83]
et al. Neuropsych. Genet./2010 patients (female/male: (three-times a as responders, the effect of the affect the response to ECT
71/33) week) COMT val158met polymorphism
on ECT response
Gedge et al. Front. Psychiatry/2012 29 TRD-MDD patients One ECT session BDNF and rTMS serum levels by ECT did not increase serum [84]
ELISA; 1 week pre- and post-ECT levels of BDNF or rTMS
Dannlowski Int. J. 104 white patients ECT treatments Ventral striatum responsiveness rs3732790, rs3773679 and [85]
et al. Neuropsychopharmacol./ with treatment- (three-times a to happy faces by means of rs9817063 variants are
2013 resistant MDD week) functional magnetic resonance associated with ECT response
(F/M: 71/33) imaging, the effect of the DRD3 and remission
polymorphism on ECT response
Viikki et al. Psychiatr. Genet./2013 119 treatment- ECT treatments Change in MADRS, testing rs11030101 is associated with [86]
Pharmacogenetics in electroconvulsive therapy & adjunctive medications

resistant MMD (three-times a the association of two BDNF the efficacy of ECT
patients week) polymorphisms, rs11030101 and
rs61888800
ECS: Electroconvulsive seizure; ECT: Electroconvulsive therapy; HAM-D: Hamilton depression rating scale; MADRS: Montgomery and Åsberg Depression Rating Scale; MDD: Major depressive disorder; OR: Odds

10.2217/PGS.15.57
Review

ratio; TRD: Treatment-resistant depression.


Review Mirzakhani, Noorden, Swen, Nozari & Guchelaar

To address the therapeutic efficacy of ECS, Newton Domschke et al., in further investigation on val­
and colleagues examined the expression of neutrophins 158met COMT, proposed that the impact of the SNP
and related signaling pathways in the hippocampus of on the efficacy of ECT in depressive patients could be
rats in response to ECS using a custom growth factor gender specific. They also suggested that the val158met
microarray chip. They reported the regulation of sev­ carrier might be less pharmacologically responsive to
eral genes that are involved in growth factor and angio­ antidepressant and could benefit from ECT in their
genic-endothelial signaling, including neuritin, stem earlier stage of mood disorder [83] .
cell factor, VEGF, (VGF), COX-2 and TIMP-1 [77] .
Some of these, as well as other identified growth fac­ DRD2 & mutual effect with COMT
tors, including VEGF, FGF and BNDF, have effects Huuhka et al. investigated the synergistic effect of
on brain neurogenesis and cell proliferation. They also two polymorphisms of DRD2 and COMT, C957T
examined gene expression in the choroid plexus and (rs6277) and Val158Met (rs4680) in response to
found several enriched growth factors in this vascular ECT treatment. The study groups comprised 118
tissue, which were affected by ECS. Among the identi­ depressive patients and 383 healthy controls [13] .
fied genes, TIMP-1 and COX-2 were highly expressed They showed that had MT Met allele and DRD2
in both acute and chronic ECS. The authors suggested T allele had synergistic effect in prediction of sever­
that the simultaneous augmented growth factor sig­ ity of depression. Furthermore, they found that the
naling with angiogenic process could have an impor­ patients with TT genotype of DRD2 C957T SNP
tant role in the mechanism underlying the therapeutic and Met/Met genotype of COMT were less likely
effect of ECT. to reach remission than those with CC genotype
of DRD2 C957T and Val/Val genotype of COMT.
Single gene approach: expression or Accordingly, they suggested the combined effect
polymorphism based of these polymorphisms might be associated with
In this approach a prior knowledge or presuppositions response to ECT [13] .
on a gene which directly or in directly may play role
in neurobiological mechanisms involved in efficacy of APOE & RGS4
ECT response was used for investigation. Huuhka et al. also examined APOE well known for
its association with neurodegenerative diseases as well
DARPP-32 as RGS4 in prediction of TRD-MDD and found no
DARPP-32 protein (dopamine- and cyclic-AMP-reg­ association between APOE and RGS4 polymorphism
ulated phosphoprotein of molecular weight 32,000) and response to ECT [69,71] . This finding was, how­
has been of interest due to its phosphorylation regula­ ever, not consistent with the only previous study on
tion by dopamine and cAMP in nerve, which might the association of APOE and ECT responders [68] .
mediate some dopamine effects [87] . DARPP-32 gene
downregulation has also been implicated in schizophe­ DRD3 (DR3)
renic patients [88] . Accordingly, Rosa et al. showed that To follow-up on the role of dopamine D receptor gene
DARPP-32 expressions in striatum and hippocampus in efficacy of ECT and evaluate the potential impact
of rats increased after five ECSs in 48 h. However, the of DRD3 variation on ECT outcome in treatment-
effect was fluctuant and transient [78] . resistant major depression, Dannlowski and colleagues
used ten genetic markers with high coverage percent­
COMT & APOE age on DRD3 to investigate the association with
COMT, a major enzyme in dopamine metabolism in response to ECT in 104 treatment-resistant MDD
the prefrontal cortex has been of interest in response white patients.
to efficacy of ECT. Anttila et al. showed that COMT They found significant association of rs3732790,
high/high genotype carriers would be more common in rs3773679 variants of DRD3 with response to ECT
responders to ECT than other genotype carriers [72] . A (p = 0.02 and 0.03, respectively) and rs9817063 SNP
finding that suggested the lower dopamine levels in the with remission (p = 0.01) after ECT. They suggested
prefrontal cortex could be associated with substantially that DRD3 gene variation might affect the efficacy
better treatment effects of ECT. COMT Val158Met, a of ECT that might potentially be mediated through
functional polymorphism of COMT at codon 158 sub­ neurobiological pathways of striatal activity [85] .
stantially affected the dopaminergic activity such that
the Met allele homozygosity resulted in considerable BDNF
reduction of enzymatic activity compared with the Val BDNF is a member of the NGF family of neuro­
allele homozygosity [72] . trophins. BDNF has been shown to exert important

10.2217/PGS.15.57 Pharmacogenomics (Epub ahead of print) future science group


Pharmacogenetics in electroconvulsive therapy & adjunctive medications Review

functions in neuronal survival, proliferation and syn­ tigated the association between BDNF polymor­
aptic plasticity in the brain [89–92] . There are several phism rs11030101 and the efficacy of ECT. Their
biological evidences to support the role of BDNF as study demonstrated that the TA genotype carriers
a central neurotrophic factor in the efficacy of ECT. of rs11030101 were less likely to show improvement
These evidences have shown the inductive effects in Montgomery-Åsberg Depression Rating Scale
of both single and repeated ECT on BDNF secre­ (MADRS) and benefit from ECT compared with
tion in brain that are reflective of BDNF changes in patients with the TT genotype [86] .
serum [55,74] . Similarly, BDNF levels are shown to
decrease in individuals with depression and increase VEGF, P2RX7 & HTR2A
following antidepressants; the changes that correlate Viikki et al. also examined the association between
with the severity of the disorder [55,93] . Accordingly, the VEGF 2578 C/A polymorphism and ECT in 119
it has been suggested that BDNF may at least in part patients with TRD who were treated with ECT and
explain both the acute and chronic potent effects of 98 depressive patients treated with SSRIs compared
ECT in depressive disorders [74,94] . with healthy controls. According to their findings,
The neurobiological mechanisms of action in ECT the CC genotype of VEGF 2578C/A polymorphism
have been proposed to be involvement in, the induc­ was more common in patients treated with ECT and
tion of BDNF secretion by prolonged increase of SSRI than in healthy controls (31.1, 25.5 and 18.7%,
both BDNF and TRKB (NTRK2) mRNA expression respectively; p = 0.056). The VEGF 2578 C/A poly­
in the hippocampus and entorhinal cortex (EC) [95] . morphism was associated with treatment-resistant
However, Taliaz et al. suggested that while neuro­ depression and CC genotype was more frequent
plastic alterations, as expressed by changes in BDNF in patients underwent ECT than in controls (31.1
expression within different brain regions, might be and 18.7%, respectively; p = 0.015) [97] . In the same
induced by ECT, the antidepressant-like effect of study groups, they investigated the rs2230912 and
ECT in an animal model depends on reduction of rs2230912 P2RX7 poly­morphisms. Neither of these
the ventral tegmental area (VTA) BDNF expres­ two P2RX7 SNP was associated with either remis­
sion but not on the elevation of hippocampal BDNF sion after SSRI or ECT [98] . In the same study popu­
expression [81] . lations, they investigated the improvement of depres­
In an animal experiment, Segawa and colleagues sion using MADRS score after ECT in association
tried to explain the role BDNF and pro-BDNF in with rs7997012 and rs6311 HTR2A polymorphisms.
acute and chronic ECT treatment. They found that None of the SNPs were associated with the change in
single administration of ECS rapidly increased hippo­ MADRS score due to treatment. However, the inter­
campal levels of pro-BDNF along with levels of PC1 action between the SNPs and gender explained 14%
and t-PA. These two proteases are involved in intra- of the variance in MADRS score change [99] .
and extra-cellular pro-BDNF processing [80] . Further
ECSs resulted in increased hippocampal level of pro- CREB
BDNF as well as mature BDNF level. Taken together, FZ6 is a seven transmembrane-spanning receptor
they suggested that while PC1 and t-PA could both involved in Wnt signaling. This signaling pathway is one
be involved in pro-BDNF processing connected with of the essential mechanisms in cell proliferation, polar­
acute antidepressant effect of ECT, t-PA might play ity and fate determination during embryonic develop­
a dominant role following repeated ECS [80] . In their ment and tissue homeostasis [100,101] . The main signal­
model, chronic administration of imipramine signifi­ ing pathway is activated by FZ/β-catenin, FZ/Ca 2+
cantly increased mature BDNF levels, but not pro- and FZ/planar cell polarity signaling pathways [102]
BDNF and protease levels, indicating that the thera­ and inhibited by Dickkopf (Dkk) family members
peutic mechanism of antidepressants might differ from (e.g., Dkk1 which functions as secreted Wnt antago­
that of ECT. nists by inhibiting Wnt coreceptors LRP5/6) [103] .
Clinical studies support some of the obtained Voleti et al. demonstrated that chronic administration
evidences by preclinical studies on BDNF. The sig­ of ECS augments the activity of several hippocampal
nificant increase in serum levels of BDNF has been genes through the CREB such that subsequent effects
detected in patients undergoing chronic ECTs [74] . might lead to the effectiveness of chronic ECT. FZ6
A possible mechanism for this observation has been was also one of CREB-target genes, which was affected
proposed to be due to BDNF gene upregulation by chronic ECS. In their study, viral vector mediated
mediated by histone H3 and H4 acetylation [96] , inhibition of Fzd6 produced anxiety and depressive-
though in preclinical setting. Viikki and colleagues, like effects [79] . Accordingly, the authors suggested that
in a recent study of 119 depressive patients, inves­ the activation of CREB might have regulatory effect

future science group 10.2217/PGS.15.57


Review Mirzakhani, Noorden, Swen, Nozari & Guchelaar

on multiple functional pathways such that the thera­ referred for ECT. All participants were genotyped for
peutic effect of ECS is dependent on a particular set of ACE, and the efficacy of ECT was evaluated using the
CREB-activated genes [79] . MADRS. ACE genotype was not associated with ECT
efficacy and did not show a different frequency with
ACE healthy controls [82] .
ACE has been suggested as a major gene affecting affec­
tive disorders and their treatment. To compare the Discussion & future perspective
effects of the ACE genotype distributions and treatment Our review demonstrates that the knowledge for safe
response to ECT in MDD patients, Stewart et al. stud­ application of ECT treatment of major depression and
ied 119 treatment-resistant depressive patients who were other psychiatric disorders has been improved, at least

IP3 intracellular PtdIns(4,5)P2 intracellular

IP3 receptor PDK (PDPK1)


PtdIns(3,4,5)P3 intracellular
1,2-Diacyglycerol intracellular
Ca(’2+) endoplasmic reticulum lumen

AKT(PKB)
PI3K cat class IA
PKC-alpha PLC-gamma 1 GRB2
PI3K reg class IA
GSK3 beta
Ca(’2+) cytosol
SOS
Axin UBC IKK (cat) H-Ras BDNF
TrkB
Tcf(Lef)
UBB Beta-catenin Shc
Ca(’2+) extracellular region Rac1
Calmodulin NGFR(TNFRSF16)
FoxD1 Collagen II
TCF7L2 (TCF4)
c-Raf-1
GCR-alpha
Lef-1 MEKK1(MAP3K1)
CaMK II NFKBIA

NMDA receptor Ubiquitin


CaMK IV c-Src MEK2(MAP2K2)
RUNX2 HDAC3HDAC2
ERK1/2 MEK1(MAP2K1)
VEGFR-2
p90Rsk Androgen receptor
TNF-alpha
MYOG JNK(MAPK8-10)
TIF1-beta c-Myc COMT
CNOT3
DDR2 SODD
p53
SMAD3
RelA (p65 NF-kB subunit) AP-1
DR3(TNFRSF12)
TWEAK(TNFSF12) CREBL2
COMTD1

E-selectin
TL1A(TNFSF15)

ERR1

Figure 2. The demonstration of regulatory pathways among BDNF, COMT, DDR2, DDR3, CREB, TRKB and NMDA
receptors in the human brain. AP-1 complex shows the most regulatory effect on the gene of interest. Green
arrows demonstrate activation and red arrows deactivation.

10.2217/PGS.15.57 Pharmacogenomics (Epub ahead of print) future science group


Pharmacogenetics in electroconvulsive therapy & adjunctive medications Review

VEGFR-2

c-Jun/Fra-1

COX-2 (PTGS2) VEGF-A


c-Jun/c-Fos

FosB/JunB
FosB
VEGFR-3
VEGF-C
JunB/Fra-1 c-Fos VEGF-D
JunB

c-Jun
JunD/c-Fos
VEGF Receptor 2 kinase inhibitor IV extracellular region
AP-1

JunD/Fra-2 ATF-2/c-Jun
JunD
VEGFR-2 inhibitors 1

VEGFR-1
JunD/Fra-1
c-Jun/Fra-1 c-Jun/Fra-2

VEGF-B

Figure 3. The demonstration of regulatory pathways among the AP-1 complex, Cox2 and VEGF in the human
brain.

partly due to the role of pharmacogenetics in applica­ experiment showed that the neurobiological mecha­
tion of anesthetic agents. Some genes such as CAC- nisms during ECT substantially differ by chronicity.
NA2D1 and CACNA1S, BCHE and RYR1 are associ­ More importantly, they showed that a subset of genes
ated with safe practice of anesthesia in ECT. While our would continue to be similarly expressed in some
review demonstrates that at this point, the knowledge brain regions by both acute and chronic ECT. This
of the mechanisms underlying the efficacy of ECT has fact could be suggestive that efficacy of ECT could
not been thoroughly elucidated, we identified several be due to some common regulatory pathways modu­
genes (i.e., BDNF, COMT, DDR2, DDR3, CREB, lated by the therapeutic stimulus. Investigating the
VEGF, COX-2, TRKB and NMDA receptor), which potential brain regulatory network pathways among
their transcriptions might play important role in treat­ the abstracted genes (BDNF, COMT, DDR2, DDR3,
ment response to ECT. It is important to identify the CREB, VEGF, COX-2 and TRKB), we identified that
neural and the molecular pathways related to these that all genes or their transmitters are co-expressed as part
might explain the mediation of the behavioral changes of transcriptionally regulatory subnetworks in brain,
by ECT and its timely application. more prominently in the frontal lobe (Figures 2 & 3) .
An ideal shock treatment produces two sets of acute In these subregulatory networks, AP-1 transcrip­
and chronic neurobiological responses that result in a tion including CREB demonstrates the most regu­
rapid and sustained treatment response for treatment latory effects on the network objects. This takes on
of psychological disorders. Accordingly, Altar et al.’s more significance that effectiveness of ECT is more

future science group 10.2217/PGS.15.57


Review Mirzakhani, Noorden, Swen, Nozari & Guchelaar

Executive summary
Pharmacogenetics & safety of electroconvulsive therapy: anesthesia adverse events
• Abnormalities in butyrylcholinesterase
–– Butyrylcholinesterase deficiency could result in a prolonged effect of the ultra-short acting depolarizing
succinylcholine, the muscle relaxant of choice of electroconvulsive therapy (ECT), due to marked decrease
in plasma cholinesterase activity.
–– Succinylcholine duration of action is prolonged in patients heterozygous for the K-variant allele, the most
frequent variant.
• Malignant hyperthermia
–– Approximately 50% of MH-related genetic variants have been found in the RYR1 gene on chromosome 19.
–– Most cases (70%) harbor one of 30 RYR1 mutations.
–– CACNA2D1 and CACNA1S variants account for less than 2% of cases and the causal genetic variant in
approximately 30% of patients is unknown.
–– The reported incidence of MH during ECT has been significantly less than other procedures requiring
general anesthesia.
• Neuroleptic malignant syndrome
–– CYP2D6 polymorphism, the enzyme through which most psychotropic drugs are metabolized and TaqIA
DRD2, a target for antipsychotic drugs, has been suggested as the link between pharmacogenetic factors
and the potential development of neuroleptic malignant syndrome.
–– In spite of these genetic links, reliable neuroleptic malignant syndrome during ECT has been reported.
Potential neurobiological mechanisms in cognitive side effects of ECT
• ECT-induced cognitive disruption might be mediated by glutamate at NMDA receptors.
–– Using ketamine as anesthetics during ECT might result in less impairment of short-term memory.
• Higher basal level of cortisol might be associated with greater degree of ECT-induced cognitive impairment.
• Acute effect of ECT causes perturbations of several amino acid transmitters and the brain biomarkers in blood
which might be associated with neuronal activity or potential neuronal injury.
Pharmacogenetics & the efficacy of ECT: effect of adjunctive psychotropic drugs on ECT outcome
• The current remission rates after ECT appear to have declined.
–– The overall remission rate has been reported to be 48.0 in 64.9% patients with and without previous
pharmacotherapy failure, respectively.
• ECT has shown higher efficacy in conjunction with antidepressants and antipsychotics.
• Application of the NMDA receptor antagonist ketamine during ECT might augment rapid and longer
antidepressant effect of ECT.
–– NMDA receptor antagonism could activate mammalian target of rapamycin (mTOR) and brain-derived
neurotrophic factor (BDNF) pathways.
Potential pharmacogenomics/neurobiological mechanisms in the efficacy of ECT
• Brain alterations in acute & chronic ECT response
–– ECT affects a wide range of brain areas.
–– Therapeutic effects on brain regions may be through structural, and/or biochemical changes.
–– ECT could have similar effect like antidepressants in downregulation of brain 5-HT 2 receptors in the limbic
and prefrontal cortical brain areas.
–– ECT could augment the increase in dopamine D2 receptors in some brain areas in patients who
received ECT.
–– ECT might effect structural plasticity within dorsal frontolimbic pathways and plasticity of white matter in
relation to therapeutic response in depression.
• Potential genes involved in ECT response and the related biological pathways: gene-expression signatures
–– Therapeutic effects of ECT might be due to mechanisms involving several amino acid transmitter changes
in the brain through overexpression of their regulatory genes.
–– There are some similarities in ECT-induced regional alterations, for example, hippocampus and frontal
lobe.
–– Several genes such as VEGF, VGF, COX-2 and TIMP-1 involved in growth factor and angiogenic-endothelial
signaling could be co-expressed by both acute and chronic ECT.
• Potential genes involved in ECT response and the related biological pathways: single gene approach:
expression or polymorphism based
–– Several individual genes have been investigated in association with ECT efficacy.
–– ECT could transiently increase DARPP-32 expressions in striatum and hippocampus.
–– The combined effect of COMT and DRD2 polymorphisms might be associated with response to ECT. COMT
high/high genotype carriers would be more common in responders to ECT than other genotype carriers.

10.2217/PGS.15.57 Pharmacogenomics (Epub ahead of print) future science group


Pharmacogenetics in electroconvulsive therapy & adjunctive medications Review

Executive summary (cont.)


Potential pharmacogenomics/neurobiological mechanisms in the efficacy of ECT (cont.)
• Potential genes involved in ECT response and the related biological pathways: single gene approach:
expression or polymorphism based (cont.)
–– DR3 rs3732790, rs3773679 SNPs are associated with remission after ECT.
–– Both single and repeated ECTs increase BDNF secretions in mRNA expression, mostly in the hippocampus
and entorhinal cortex.
–– The BDNF change in brain is reflective of changes in serum.
–– PC1 and t-PA could both be involved in BDNF expression processing connected with acute
antidepressant effect of ECT; t-PA might play a dominant role following repeated electro­convulsive
seizure.
–– TA genotype carriers of rs11030101 were shown to be less likely to show improvement in Montgomery-
Åsberg Depression Rating Scale (MADRS) and benefit from ECT compared with patients with the TT
genotype.
• VEGF 2578 CC genotype was observed with more frequency in patients who underwent ECT.
• CREB might have regulatory effect on multiple functional pathways such that the therapeutic effect of
electro­convulsive seizure is dependent on a particular set of CREB-activated genes.
Discussion & future perspective
• BDNF, COMT, DDR2, DDR3, CREB, VEGF, COX-2, TRKB and NMDA receptors are genes whose functions could
affect the efficacy of ECT.
• The potential genes involved in efficacy of the ECT or their transmitters are co-expressed as part of
transcriptionally regulatory subnetworks.
• In these subregulatory networks, AP-1 transcription including CREB could be a major regulator of the network
objects.
• AP-1 complex has shown high expression by administration of chronic ECT and persists to be highly expressed
by 7 days after the last ECT.
• The shared regulatory pathways such as AP-1 and CREB could be useful for further investigation to identify
novel gene therapeutic targets for treatment-resistant psychological disease.
• Investigating ECT signature genes as compared with different drug compounds could help in identifying
medications that might augment the induced differential gene expression by ECT.
• Further well-designed longitudinal clinical studies are required to increase our knowledge of the mechanisms
underlying the efficacy of ECT.

dependent on treatment response to its chronic appli­ more cumbersome than medications with higher effi­
cation. Consistently, Hope et al. [104] showed AP-1 cacy. Accordingly, our understanding of the acute and
complex shows high expression by administration of chronic molecular, cellular and behavioral changes by
chronic ECT and persists to be highly expressed by 7 ECT will provide a new view to find potential tar­
days after the last ECT. It has been shown that AP-1 gets for novel psychotropic treatments, particularly
modulation affects many cell processes including cell antidepressants, that are highlighted by the findings
proliferation, differentiation, transformation, neuro­ such as regional gene induction (e.g., BNDF, Cox-2),
nal activity and growth factor signaling [105,106] . More increased neurogenesis, electrophysiological reactiv­
importantly, it has been suggested that AP-1 could act ity, the role of VEGF in neurogenesis [108,109] and
as an environmental biosensor in mediating the linked DARPP-32 expression [78] . Some genes and associated
cellular biological process [105] . pathways such as BDNF, TRKB-MAP kinase path­
There are other factors that might affect the varia­ way, NPY, VEGF, arachidonic acid pathway, TRH,
tions in expression levels in brain regions and direc­ VEGF and neurogenesis regulation pathways, which
tionality of regional regulatory networks. For exam­ have shown differential expressions due to chronic
ple, there is evidence of variations in activity level of stimulation by ECT, are more probable to have an
BDNF transcripts in different brain regions [81,107] or intermediary role in benefiting the long-term effects
other patients’ clinical characteristics such as gender of ECT. These evidences along with shared regulatory
might influence some observed gene polymorphisms pathways such as AP-1 and CREB could be useful for
in response to treatment [99] . further investigation to identify novel gene targets for
In spite of all the improved knowledge on the treating treatment-resistant psychological disease.
safety of ECT and its proved efficacy in treatment Palfreyman et al. suggest illustrating a ‘disease signa­
of some psychiatric patients unresponsive to medi­ ture’ and ‘drug signature’ of aberrantly expressed genes
cal therapy, ECT is still a physical intervention and from comparison of normal controls and patients [110] .

future science group 10.2217/PGS.15.57


Review Mirzakhani, Noorden, Swen, Nozari & Guchelaar

In this approach, the genes associated with disease will could also be used to augment the induced differential
be explored. The comparison of the identified genes genes expressions by chronic exposure to ECT or by
with ECT signature genes could identify a set of tar­ anti­psychotics [55] . Further well-designed longitudinal
gets whose alteration might be a better predictor of dis­ clinical studies are required to increase our knowledge
ease and the effect of procedural treatment. Ultimately, of the mechanisms underlying the efficacy of ECT.
such overlapping genes could be used to identify drug
compounds that show similarity in inducing the gene Financial & competing interests disclosure
expressions, which consequently mimic the therapeu­ The authors have no relevant affiliations or financial involve-
tic response of ECT. This method has been applied ment with any organization or entity with a financial inter-
in animal mode such that [111] several compounds est in or financial conflict with the subject matter or mate-
have been investigated to identify the one that alter rials discussed in the manuscript. This includes employment,
the same 11 genes elevated by ECT or exercise in rat consultancies, honoraria, stock ownership or options, expert
brain. Of note, exercise has been favorably compared testimony, grants or patents received or pending, or royalties.
with antidepressant treatment for treatment of mild- No writing assistance was utilized in the production of this
to-moderate depression [111–113] . Such compounds manuscript.

References analysis of 83 patients. J. Clin. Psychiatry 70(1), 92–94


(2009).
1 Mirzakhani H, Welch CA, Eikermann M, Nozari A.
Neuromuscular blocking agents for electroconvulsive 13 Huuhka K, Anttila S, Huuhka M et al. Dopamine 2 receptor
therapy: a systematic review. Acta Anaesthesiol. Scand. 56(1), C957T and catechol-O-methyltransferase Val158Met
3–16 (2012). polymorphisms are associated with treatment response in
electroconvulsive therapy. Neurosci. Lett. 448(1), 79–83
2 Fochtmann LJ. Genetic approaches to the neurobiology of
(2008).
electroconvulsive therapy. J. ECT 14(3), 206–219 (1998).
14 Bauld HW, Gibson PF, Jebson PJ, Brown SS. Aetiology
3 Kalow W. Atypical plasma cholinesterase. A personal
of prolonged apnoea after suxamethonium. Br. J. Anaesth.
discovery story: a tale of three cities. Can. J. Anaesth. 51(3),
46(4), 273–281 (1974).
206–211 (2004).
15 Levano S, Keller D, Schobinger E, Urwyler A, Girard T.
4 Kalow W, Staron N. On distribution and inheritance of
Rapid and accurate detection of atypical and Kalow variants
atypical forms of human serum cholinesterase, as indicated
in the butyrylcholinesterase gene using denaturing high
by dibucaine numbers. Can. J. Biochem. Physiol. 35(12),
performance liquid chromatography. Anesthesia Analgesia
1305–1320 (1957).
106(1), 147–151 (2008).
5 Kalow W. Pharmacogenetics: a historical perspective.
16 Yen T, Nightingale BN, Burns JC, Sullivan DR, Stewart
In: Pharmacogenomics: Applications to Patient Care. Allen
PM. Butyrylcholinesterase (BCHE) genotyping for post-
WL JJ, Knoell DL et al. (Eds). American College of Clinical
succinylcholine apnea in an Australian population. Clin.
Pharmacy, Kansas City, MO, USA, 251–269 (2004).
Chem. 49(8), 1297–1308 (2003).
6 Roden DM, Altman RB, Benowitz NL et al.
17 Jensen FS, Viby-Mogensen J. Plasma cholinesterase and
Pharmacogenomics: challenges and opportunities. Ann.
abnormal reaction to succinylcholine: twenty years’
Intern. Med. 145(10), 749–757 (2006).
experience with the Danish Cholinesterase Research Unit.
7 Galley HF, Mahdy A, Lowes DA. Pharmacogenetics and Acta Anaesthesiol. Scand. 39(2), 150–156 (1995).
anesthesiologists. Pharmacogenomics 6(8), 849–856 (2005).
18 Zavorotnyy M, Zwanzger P. Prolonged apnea during
8 Jelovac A, Kolshus E, McLoughlin DM. Relapse following electroconvulsive therapy in monozygotic twins: case reports.
successful electroconvulsive therapy for major depression: Ann. Gen. Psychiatry 10(1), 30 (2011).
a meta-analysis. Neuropsychopharmacology 38(12), 2467–2474
19 Omprakash TM, Surender P. Prolonged apnea following
(2013).
modified electroconvulsive therapy with suxamethonium.
9 Tharyan P, Adams CE. Electroconvulsive therapy for Indian J. Psychol. Med. 33(2), 191–193 (2011).
schizophrenia. Cochrane Database Syst. Rev. 2, CD000076
20 Waghmare A, Kumar CN, Thirthalli J. Suxamethonium
(2005).
induced prolonged apnea in a patient receiving
10 Group UER. Efficacy and safety of electroconvulsive therapy electroconvulsive therapy. General Hospital Psychiatry 32(4),
in depressive disorders: a systematic review and meta- 447 e441–e442 (2010).
analysis. Lancet 361(9360), 799–808 (2003).
21 Mollerup HM, Gatke MR. Butyrylcholinesterase
11 Wilkins KM, Ostroff R, Tampi RR. Efficacy of gene mutations in patients with prolonged apnea after
electroconvulsive therapy in the treatment of nondepressed succinylcholine for electroconvulsive therapy. Acta
psychiatric illness in elderly patients: a review of the Anaesthesiol. Scand. 55(1), 82–86 (2011).
literature. J. Geriatr. Psychiatry Neurol. 21(1), 3–11 (2008).
22 Rice J, Lebowitz PW, Bailine SH, Mowerman S. Malignant
12 Rasmussen KG, Black JL. Serotonin transporter gene status hyperthermia and electroconvulsive therapy. Convulsive Ther.
and electroconvulsive therapy outcomes: a retrospective 9(1), 45–49 (1993).

10.2217/PGS.15.57 Pharmacogenomics (Epub ahead of print) future science group


Pharmacogenetics in electroconvulsive therapy & adjunctive medications Review

23 Pearlman C. Neuroleptic malignant syndrome, malignant 41 Huuhka K, Viikki M, Tammentie T et al. One-year follow-
hyperthermia, and ECT. Convulsive Ther. 4(2), 181 (1988). up after discontinuing maintenance electroconvulsive
24 Yacoub OF, Morrow DH. Malignant hyperthermia and therapy. J. ECT 28(4), 225–228 (2012).
ECT. Am. J. Psychiatry 143(8), 1027–1029 (1986). 42 Heijnen WT, Birkenhager TK, Wierdsma AI, van den
25 Brandom BW. Genetics of malignant hyperthermia. Scientific Broek WW. Antidepressant pharmacotherapy failure and
World J. 6, 1722–1730 (2006). response to subsequent electroconvulsive therapy: a meta-
analysis. J. Clin. Psychopharmacol. 30(5), 616–619 (2010).
26 Brandom BW, Bina S, Wong CA et al. Ryanodine receptor
type 1 gene variants in the malignant hyperthermia- 43 Haskett RF, Loo C. Adjunctive psychotropic medications
susceptible population of the United States. Anesthesia during electroconvulsive therapy in the treatment of
Analgesia 116(5), 1078–1086 (2013). depression, mania, and schizophrenia. J. ECT 26(3),
196–201 (2010).
27 Robinson R, Carpenter D, Shaw MA, Halsall J, Hopkins P.
Mutations in RYR1 in malignant hyperthermia and central 44 Zhang JP, Malhotra AK. Pharmacogenetics and
core disease. Hum. Mutat. 27(10), 977–989 (2006). antipsychotics: therapeutic efficacy and side effects
prediction. Expert Opin. Drug Metab. Toxicol. 7(1), 9–37
28 Gronert GA. Malignant hyperthermia. Anesthesiology 53(5),
(2011).
395–423 (1980).
45 Murrough JW, Iosifescu DV, Chang LC et al.
29 Johnson GC, Santos AB. More on ECT and malignant
Antidepressant efficacy of ketamine in treatment-resistant
hyperthermia. Am. J. Psychiatry 140(2), 266–267 (1983).
major depression: a two-site randomized controlled trial.
30 Zivkovic M, Mihaljevic-Peles A, Sagud M, Silic A, Am. J. Psychiatry 170(10), 1134–1142 (2013).
Mihanovic M. The role of CYP2D6 and TaqI A
46 Murrough JW, Perez AM, Pillemer S et al. Rapid and
polymorphisms in malignant neuroleptic syndrome: two case
longer-term antidepressant effects of repeated ketamine
reports with three episodes. Psychiatr Danub. 22(1), 112–116
infusions in treatment-resistant major depression. Biol.
(2010).
Psychiatry 74(4), 250–256 (2013).
31 Trollor JN, Sachdev PS. Electroconvulsive treatment of
47 Berman RM, Cappiello A, Anand A et al. Antidepressant
neuroleptic malignant syndrome: a review and report of
effects of ketamine in depressed patients. Biol. Psychiatry
cases. Aust. NZ J. Psychiatry 33(5), 650–659 (1999).
47(4), 351–354 (2000).
32 Davis JM, Janicak PG, Sakkas P, Gilmore C, Wang Z.
48 Diazgranados N, Ibrahim L, Brutsche NE et al.
Electroconvulsive therapy in the treatment of the neuroleptic
A randomized add-on trial of an N-methyl-D-aspartate
malignant syndrome. Convulsive Ther. 7(2), 111–120 (1991).
antagonist in treatment-resistant bipolar depression. Arch.
33 Nobler MS, Sackeim HA. Neurobiological correlates of the Gen. Psychiatry 67(8), 793–802 (2010).
cognitive side effects of electroconvulsive therapy. J. ECT
49 Zarate CA Jr, Brutsche NE, Ibrahim L et al. Replication
24(1), 40–45 (2008).
of ketamine’s antidepressant efficacy in bipolar depression:
34 Khan A, Mirolo MH, Mirolo HA, Miller S. Can ECT- a randomized controlled add-on trial. Biol. Psychiatry
induced cognitive effects be altered pharmacologically? 71(11), 939–946 (2012).
Prog. Neuropsychopharmacol. Biol. Psychiatry 17(6), 861–873
50 Carlson PJ, Diazgranados N, Nugent AC et al. Neural
(1993).
correlates of rapid antidepressant response to ketamine
35 Lerer B. Studies on the role of brain cholinergic systems in in treatment-resistant unipolar depression: a preliminary
the therapeutic mechanisms and adverse effects of ECT and positron emission tomography study. Biol. Psychiatry
lithium. Biol. Psychiatry 20(1), 20–40 (1985). 73(12), 1213–1221 (2013).
36 Prudic J, Fitzsimons L, Nobler MS, Sackeim HA. 51 Murrough JW. Ketamine as a novel antidepressant:
Naloxone in the prevention of the adverse cognitive from synapse to behavior. Clin. Pharmacol. Ther. 91(2),
effects of ECT: a within-subject, placebo controlled study. 303–309 (2012).
Neuropsychopharmacology 21(2), 285–293 (1999).
52 Wang X, Chen Y, Zhou X, Liu F, Zhang T, Zhang C.
37 McDaniel WW, Sahota AK, Vyas BV, Laguerta N, Hategan Effects of propofol and ketamine as combined anesthesia
L, Oswald J. Ketamine appears associated with better word for electroconvulsive therapy in patients with depressive
recall than etomidate after a course of 6 electroconvulsive disorder. J. ECT 28(2), 128–132 (2012).
therapies. J. ECT 22(2), 103–106 (2006).
53 Abdallah CG, Fasula M, Kelmendi B, Sanacora G,
38 Neylan TC, Canick JD, Hall SE, Reus VI, Sapolsky RM, Ostroff R. Rapid antidepressant effect of ketamine in the
Wolkowitz OM. Cortisol levels predict cognitive impairment electroconvulsive therapy setting. J. ECT 28(3), 157–161
induced by electroconvulsive therapy. Biol. Psychiatry 50(5), (2012).
331–336 (2001).
54 Fochtmann LJ. Animal studies of electroconvulsive
39 Palmio J, Huuhka M, Laine S et al. Electroconvulsive therapy: foundations for future research. Psychopharmacol.
therapy and biomarkers of neuronal injury and plasticity: Bull. 30(3), 321–444 (1994).
serum levels of neuron-specific enolase and S-100b protein.
55 Altar CA, Laeng P, Jurata LW et al. Electroconvulsive
Psychiatry Res. 177(1–2), 97–100 (2010).
seizures regulate gene expression of distinct neurotrophic
40 Palmio J, Huuhka M, Saransaari P et al. Changes in plasma signaling pathways. J. Neurosci. 24(11), 2667–2677
amino acids after electroconvulsive therapy of depressed (2004).
patients. Psychiatry Res. 137(3), 183–190 (2005).

future science group 10.2217/PGS.15.57


Review Mirzakhani, Noorden, Swen, Nozari & Guchelaar

56 Yatham LN, Liddle PF, Erez J et al. Brain serotonin-2 71 Huuhka K, Kampman O, Anttila S et al. RGS4
receptors in acute mania. Br. J. Psychiatry 196(1), 47–51 polymorphism and response to electroconvulsive therapy
(2010). in major depressive disorder. Neurosci. Lett. 437(1), 25–28
57 Yatham LN, Liddle PF, Lam RW et al. Effect of (2008).
electroconvulsive therapy on brain 5-HT(2) receptors in 72 Anttila S, Huuhka K, Huuhka M et al. Catechol-O-
major depression. Br. J. Psychiatry 196(6), 474–479 (2010). methyltransferase (COMT) polymorphisms predict
58 Lanzenberger R, Baldinger P, Hahn A et al. Impact of treatment response in electroconvulsive therapy.
electroconvulsive therapy on 5-HT1A receptor binding in Pharmacogenomics J. 8(2), 113–116 (2008).
major depression. Mol. Psychiatry 18(1), 1 (2013). 73 Anttila S, Huuhka K, Huuhka M et al. Interaction between
59 Saijo T, Takano A, Suhara T et al. Effect of electroconvulsive TPH1 and GNB3 genotypes and electroconvulsive therapy
therapy on 5-HT1A receptor binding in patients with in major depression. J. Neural Transm. 114(4), 461–468
depression: a PET study with [11C]WAY 100635. Int. (2007).
J. Neuropsychopharmacol. 13(6), 785–791 (2010). 74 Bocchio-Chiavetto L, Zanardini R, Bortolomasi M et al.
60 Saijo T, Takano A, Suhara T et al. Electroconvulsive therapy Electroconvulsive Therapy (ECT) increases serum brain
decreases dopamine D(2)receptor binding in the anterior derived neurotrophic factor (BDNF) in drug resistant
cingulate in patients with depression: a controlled study depressed patients. Eur. Neuropsychopharmacol. 16(8),
using positron emission tomography with radioligand [(1)(1) 620–624 (2006).
C]FLB 457. J. Clin. Psychiatry 71(6), 793–799 (2010). 75 Nibuya M, Morinobu S, Duman RS. Regulation of BDNF
61 Drevets WC, Price JL, Furey ML. Brain structural and and trkB mRNA in rat brain by chronic electroconvulsive
functional abnormalities in mood disorders: implications seizure and antidepressant drug treatments. J. Neurosci.
for neurocircuitry models of depression. Brain Struct. Funct. 15(11), 7539–7547 (1995).
213(1–2), 93–118 (2008). 76 Zetterstrom TS, Pei Q, Grahame-Smith DG. Repeated
62 Liao Y, Huang X, Wu Q et al. Is depression a disconnection electroconvulsive shock extends the duration of enhanced
syndrome? Meta-analysis of diffusion tensor imaging studies gene expression for BDNF in rat brain compared with a
in patients with MDD. J. Psychiatry Neurosci. 38(1), 49–56 single administration. Brain Res. Mol. Brain Res. 57(1),
(2013). 106–110 (1998).

63 Mayberg HS. Modulating dysfunctional limbic-cortical 77 Newton SS, Collier EF, Hunsberger J et al. Gene profile of
circuits in depression: towards development of brain-based electroconvulsive seizures: induction of neurotrophic and
algorithms for diagnosis and optimised treatment. Br. Med. angiogenic factors. J. Neurosci. 23(34), 10841–10851 (2003).
Bull. 65, 193–207 (2003). 78 Rosa DV, Souza RP, Souza BR et al. DARPP-32 expression in
64 Cole J, Chaddock CA, Farmer AE et al. White matter rat brain after electroconvulsive stimulation. Brain Res. 1179,
abnormalities and illness severity in major depressive 35–41 (2007).
disorder. Br. J. Psychiatry 201(1), 33–39 (2012). 79 Voleti B, Tanis KQ, Newton SS, Duman RS. Analysis of
65 de Diego-Adelino J, Pires P, Gomez-Anson B et al. target genes regulated by chronic electroconvulsive therapy
Microstructural white-matter abnormalities associated with reveals role for Fzd6 in depression. Biol. Psychiatry 71(1),
treatment resistance, severity and duration of illness in major 51–58 (2012).
depression. Psychol. Med. 44(6), 1171–1182 (2014). 80 Segawa M, Morinobu S, Matsumoto T, Fuchikami M,
66 Hoogenboom WS, Perlis RH, Smoller JW et al. Limbic Yamawaki S. Electroconvulsive seizure, but not imipramine,
system white matter microstructure and long-term treatment rapidly up-regulates pro-BDNF and t-PA, leading to
outcome in major depressive disorder: a diffusion tensor mature BDNF production, in the rat hippocampus. Int.
imaging study using legacy data. World J. Biol. Psychiatry J. Neuropsychopharmacol. 16(2), 339–350 (2013).
15(2), 122–134 (2014). 81 Taliaz D, Nagaraj V, Haramati S, Chen A, Zangen A. Altered
67 Taylor WD, Kuchibhatla M, Payne ME et al. Frontal white brain-derived neurotrophic factor expression in the ventral
matter anisotropy and antidepressant remission in late-life tegmental area, but not in the hippocampus, is essential for
depression. PLoS ONE 3(9), e3267 (2008). antidepressant-like effects of electroconvulsive therapy. Biol.
Psychiatry 74(4), 305–312 (2013).
68 Fisman M, Rabheru K, Hegele RA et al. Apolipoprotein E
polymorphism and response to electroconvulsive therapy. 82 Stewart JA, Kampman O, Huuhka M et al. ACE
J. ECT 17(1), 11–14 (2001). polymorphism and response to electroconvulsive therapy in
major depression. Neurosci. Lett. 458(3), 122–125 (2009).
69 Huuhka M, Anttila S, Leinonen E et al. The apolipoprotein
E polymorphism is not associated with response to 83 Domschke K, Zavorotnyy M, Diemer J et al. COMT
electroconvulsive therapy in major depressive disorder. val158met influence on electroconvulsive therapy response
J. ECT 21(1), 7–11 (2005). in major depression. Am. J. Med. Genet. Part B, Neuropsych.
Genet. 153B(1), 286–290 (2010).
70 Huuhka K, Anttila S, Huuhka M et al. Brain-derived
neurotrophic factor (BDNF) polymorphisms G196A and 84 Gedge L, Beaudoin A, Lazowski L, du Toit R, Jokic R,
C270T are not associated with response to electroconvulsive Milev R. Effects of electroconvulsive therapy and repetitive
therapy in major depressive disorder. Eur. Arch. Psychiatry transcranial magnetic stimulation on serum brain-derived
Clin. Neurosci. 257(1), 31–35 (2007). neurotrophic factor levels in patients with depression. Front.
Psychiatry 3, 12 (2012).

10.2217/PGS.15.57 Pharmacogenomics (Epub ahead of print) future science group


Pharmacogenetics in electroconvulsive therapy & adjunctive medications Review

85 Dannlowski U, Domschke K, Birosova E et al. Dopamine D(3) 99 Viikki M, Huuhka K, Leinonen E et al. Interaction between
receptor gene variation: impact on electroconvulsive therapy two HTR2A polymorphisms and gender is associated with
response and ventral striatum responsiveness in depression. Int. treatment response in MDD. Neurosci. Lett. 501(1), 20–24
J. Neuropsychopharmacol. 16(7), 1443–1459 (2013). (2011).
86 Viikki ML, Jarventausta K, Leinonen E et al. BDNF 100 MacDonald BT, Tamai K, He X. Wnt/beta-catenin
polymorphism rs11030101 is associated with the efficacy of signaling: components, mechanisms, and diseases.
electroconvulsive therapy in treatment-resistant depression. Developmental Cell 17(1), 9–26 (2009).
Psychiatr. Genet. 23(3), 134–136 (2013). 101 Komiya Y, Habas R. Wnt signal transduction pathways.
87 Hemmings HC Jr, Greengard P, Tung HY, Cohen P. Organogenesis 4(2), 68–75 (2008).
DARPP-32, a dopamine-regulated neuronal phosphoprotein, is 102 Seifert JR, Mlodzik M. Frizzled/PCP signalling: a conserved
a potent inhibitor of protein phosphatase-1. Nature 310(5977), mechanism regulating cell polarity and directed motility.
503–505 (1984). Nat. Rev. Genet. 8(2), 126–138 (2007).
88 Yger M, Girault JA. DARPP-32, jack of all trades. master of 103 Niehrs C. Function and biological roles of the Dickkopf
which? Front. Behav. Neurosci. 5, 56 (2011). family of Wnt modulators. Oncogene 25(57), 7469–7481
89 Lindholm D, Castren E, Berzaghi M, Blochl A, Thoenen H. (2006).
Activity-dependent and hormonal regulation of neurotrophin 104 Hope BT, Kelz MB, Duman RS, Nestler EJ. Chronic
mRNA levels in the brain—implications for neuronal plasticity. electroconvulsive seizure (ECS) treatment results in
J. Neurobiol. 25(11), 1362–1372 (1994). expression of a long-lasting AP-1 complex in brain with
90 Binder DK, Scharfman HE. Brain-derived neurotrophic factor. altered composition and characteristics. J. Neurosci. 14(7),
Growth Factors 22(3), 123–131 (2004). 4318–4328 (1994).
91 Kocabas NA, Antonijevic I, Faghel C et al. Brain-derived 105 Wagner EF. AP-1 – introductory remarks. Oncogene 20(19),
neurotrophic factor gene polymorphisms: influence on 2334–2335 (2001).
treatment response phenotypes of major depressive disorder. 106 Kaminska B, Pyrzynska B, Ciechomska I, Wisniewska
Int. Clin. Psychopharmacol. 26(1), 1–10 (2011). M. Modulation of the composition of AP-1 complex and
92 Conti B, Maier R, Barr AM et al. Region-specific its impact on transcriptional activity. Acta Neurobiol. Exp.
transcriptional changes following the three antidepressant (Wars) 60(3), 395–402 (2000).
treatments electro convulsive therapy, sleep deprivation and 107 Pruunsild P, Kazantseva A, Aid T, Palm K, Timmusk
fluoxetine. Mol. Psychiatry 12(2), 167–189 (2007). T. Dissecting the human BDNF locus: bidirectional
93 Brunoni AR, Lopes M, Fregni F. A systematic review and meta- transcription, complex splicing, and multiple promoters.
analysis of clinical studies on major depression and BDNF Genomics 90(3), 397–406 (2007).
levels: implications for the role of neuroplasticity in depression. 108 Segi-Nishida E. Exploration of new molecular mechanisms
Int. J. Neuropsychopharmacol. 11(8), 1169–1180 (2008). for antidepressant actions of electroconvulsive seizure. Biol.
94 Kampman O, Leinonen E. Efficacy of electroconvulsive Pharm. Bull. 34(7), 939–944 (2011).
therapy: is it in the BDNF gene? Pharmacogenomics 14(12), 109 Segi-Nishida E, Warner-Schmidt JL, Duman RS.
1365–1368 (2013). Electroconvulsive seizure and VEGF increase the
95 Madsen TM, Treschow A, Bengzon J, Bolwig TG, Lindvall proliferation of neural stem-like cells in rat hippocampus.
O, Tingstrom A. Increased neurogenesis in a model of Proc. Natl Acad. Sci. USA 105(32), 11352–11357 (2008).
electroconvulsive therapy. Biol. Psychiatry 47(12), 1043–1049 110 Palfreyman MG, Hook DJ, Klimczak LJ, Brockman JA,
(2000). Evans DM, Altar CA. Novel directions in antipsychotic
96 Tsankova NM, Kumar A, Nestler EJ. Histone modifications target identification using gene arrays. Curr. Drug Targets
at gene promoter regions in rat hippocampus after acute CNS Neurol. Disord. 1(2), 227–238 (2002).
and chronic electroconvulsive seizures. J. Neurosci. 24(24), 111 Hill RD, Storandt M, Malley M. The impact of long-term
5603–5610 (2004). exercise training on psychological function in older adults.
97 Viikki M, Anttila S, Kampman O et al. Vascular endothelial J. Gerontol. 48(1), P12–P17 (1993).
growth factor (VEGF) polymorphism is associated with 112 Labbe EE, Welsh MC, Delaney D. Effects of consistent
treatment resistant depression. Neurosci. Lett. 477(3), 105–108 aerobic exercise on the psychological functioning of women.
(2010). Percept Mot. Skills. 67(3), 919–925 (1988).
98 Viikki M, Kampman O, Anttila S et al. P2RX7 113 Carek PJ, Laibstain SE, Carek SM. Exercise for the treatment
polymorphisms Gln460Arg and His155Tyr are not associated of depression and anxiety. Int. J. Psychiatry Med. 41(1),
with major depressive disorder or remission after SSRI or ECT. 15–28 (2011).
Neurosci. Lett. 493(3), 127–130 (2011).

future science group 10.2217/PGS.15.57

You might also like