You are on page 1of 19

Progress in Neuropsychopharmacology & Biological Psychiatry 121 (2023) 110672

Contents lists available at ScienceDirect

Progress in Neuropsychopharmacology & Biological


Psychiatry
journal homepage: www.elsevier.com/locate/pnp

Transcranial direct current stimulation for bipolar depression: systematic


reviews of clinical evidence and biological underpinnings
Giordano D'Urso a, *, Elena Toscano a, Annarita Barone a, Mario Palermo a,
Bernardo Dell'Osso b, f, g, Giorgio Di Lorenzo c, h, i, Antonio Mantovani d, j, Giovanni Martinotti e, k,
Michele Fornaro a, Felice Iasevoli a, Andrea de Bartolomeis a
a
Section of Psychiatry, Clinical Unit of Psychiatry and Psychology, Unit of Treatment Resistance in Psychiatry, Laboratory of Neuromodulation, Laboratory of Molecular
and Translational Psychiatry, Department of Neurosciences, Reproductive and Odontostomatological Sciences, Clinical Department of Head and Neck, University of
Naples Federico II, Napoli, Italy
b
Department of Biomedical and Clinical Sciences Luigi Sacco, Ospedale Luigi Sacco Polo Universitario, ASST Fatebenefratelli Sacco, Milan, Italy
c
Laboratory of Psychophysiology and Cognitive Neuroscience, Department of Systems Medicine, Tor Vergata University of Rome, Italy
d
Dipartimento di Medicina e Scienze della Salute "V. Tiberio" Università degli Studi del Molise, Campobasso, Italy
e
Department of Neuroscience, Imaging, Clinical Sciences, University Gabriele d’Annunzio, Chieti-Pescara, Italy
f
Department of Psychiatry and Behavioural Sciences, Bipolar Disorders Clinic, Stanford University, CA, USA
g
CRC "Aldo Ravelli" for Neuro-technology & Experimental Brain Therapeutics, University of Milan, Italy
h
Psychiatric and Clinical Psychology Unit, Fondazione Policlinico Tor Vergata, Rome, Italy
i
IRCCS Fondazione Santa Lucia, Rome, Italy
j
Dipartimento di Salute Mentale e delle Dipendenze, Azienda Sanitaria Regionale del Molise (ASReM), Campobasso, Italy
k
Department of Pharmacy, Pharmacology, Clinical Sciences, University of Hertfordshire, Herts, UK

A R T I C L E I N F O A B S T R A C T

Keywords: Despite multiple available treatments for bipolar depression (BD), many patients face sub-optimal responses.
Transcranial direct current stimulation (tDCS) Transcranial direct current stimulation (tDCS) has been advocated in the management of different conditions,
Transcranial magnetic stimulation (TMS) including BD, especially in treatment-resistant cases. The optimal dose and timing of tDCS, the mutual influence
Bipolar disorder
with other concurrently administered interventions, long-term efficacy, overall safety, and biological un­
Psychotic depression
Dopamine
derpinnings nonetheless deserve additional assessment. The present study appraised the existing clinical evi­
Serotonin dence about tDCS for bipolar depression, delving into the putative biological underpinnings with a special
Glutamate emphasis on cellular and molecular levels, with the ultimate goal of providing a translational perspective on the
Synaptic plasticity matter. Two separate systematic reviews across the PubMed database since inception up to August 8th 2022 were
performed, with fourteen clinical and nineteen neurobiological eligible studies. The included clinical studies
encompass 207 bipolar depression patients overall and consistently document the efficacy of tDCS, with a
reduction in depression scores after treatment ranging from 18% to 92%. The RCT with the largest sample clearly
showed a significant superiority of active stimulation over sham. Mild-to-moderate and transient adverse effects
are attributed to tDCS across these studies. The review of neurobiological literature indicates that several mo­
lecular mechanisms may account for the antidepressant effect of tDCS in BD patients, including the action on
calcium homeostasis in glial cells, the enhancement of LTP, the regulation of neurotrophic factors and inflam­
matory mediators, and the modulation of the expression of plasticity-related genes. To the best of our knowledge,
this is the first study on the matter to concurrently provide a synthesis of the clinical evidence and an in-depth
appraisal of the putative biological underpinnings, providing consistent support for the efficacy, safety, and
tolerability of tDCS.

* Corresponding author at; Section of Psychiatry, Clinical Unit of Psychiatry and Psychology, Unit of Treatment Resistance in Psychiatry, Laboratory of Neuro­
modulation, Department of Neurosciences, Reproductive and Odontostomatological Sciences, Clinical Department of Head and Neck, University of Naples “Federico
II”, Via Pansini 5, Building 18, 3rd floor, 80131 Naples, Italy.
E-mail address: giordano.durso@unina.it (G. D'Urso).

https://doi.org/10.1016/j.pnpbp.2022.110672
Received 19 May 2022; Received in revised form 9 October 2022; Accepted 26 October 2022
Available online 1 November 2022
0278-5846/© 2022 Elsevier Inc. All rights reserved.
G. D'Urso et al. Progress in Neuropsychopharmacology & Biological Psychiatry 121 (2023) 110672

1. Introduction via a long-term potentiation (LTP)-like process (Rroji et al., 2015). TDCS
studies involving patients with unipolar depression (Brunoni et al.,
Bipolar disorder (BD) is a severe condition (Merikangas et al., 2011; 2016) employed a bifrontal montage with the excitatory electrode
Soares-Weiser et al., 2007) accounting for a significant burden to the (anode) placed over the left DLPFC (F3 in the 10-20 EEG system), and
suffering one in terms of suicidal risk and overall functioning (Ferrari the inhibitory electrode (cathode) over the right DLPFC (F4), or right
et al., 2016; Phillips and Kupfer, 2013). Depressive episodes represent lateral orbitofrontal (F8), or right supraorbital cortex (Fp2). The ratio­
the prominent mood polarity of BD in most cases (Suppes et al., 2005). nale for these electrode positionings is that functional imaging studies
Except for electroconvulsive therapy (ECT) – which remains largely showed a prefrontal asymmetry in unipolar depressed patients, with
underutilized for several reasons in some geographical regions (Buccelli hypoactivity in the left dorsolateral prefrontal cortex (DLPFC) (Kennedy
et al., 2016; Cattaneo et al., 2022) – the available treatment options for et al., 1997) and relative hyperactivity in the right DLPFC (Grimm et al.,
bipolar depression are limited and often unsatisfactory, as well as 2008).
possibly leading to considerable side effects including, but not limited However, no evidence is available to inform the choice of electrode
to, affective switch, induction of rapid-cycling course, and increased risk montage and other stimulation parameters (e.g., current intensity,
for suicidality (Grunze et al., 2013; Vieta et al., 2010). In particular, no number, and duration of sessions) based on the specific mechanisms of
significant benefit vs. placebo or active control was found for antide­ bipolar depression. In fact, unipolar and bipolar disorders have complex,
pressants in terms of response and remission (Sidor and MacQueen, distinctive neurobiological underpinnings, being polygenic and multi­
2012). Among anticonvulsant mood-stabilizers, only lamotrigine led to factorial (Akula et al., 2016; Kerner, 2015), that account for dysfunc­
higher rates of treatment response vs. placebo (Geddes et al., 2009). tional synaptic plasticity and cortical-subcortical brain network
Second-generation antipsychotics would significantly improve acute dysregulation (Rodríguez-Cano et al., 2017). Despite advances in un­
bipolar depression with mixed features (Fornaro et al., 2016), especially derstanding putative pathophysiology and neurobiology (Akula et al.,
for lurasidone (Fornaro et al., 2017) and cariprazine (De Berardis et al., 2016; Tomasetti et al., 2017), omics (MacDonald et al., 2019), and
2016). Lithium, despite being the cornerstone treatment of BD, would functional neuroanatomy of bipolar disorders (Miller and Kelsoe, 2017),
not even suffice as a standalone treatment for the management of acute the specific molecular pathogenesis of the depressive polarity is still
bipolar depression. In one study, intensive psychotherapy was shown to elusive, also not fully understood are the cellular and molecular mech­
induce significantly higher year-end recovery rates, shorter time to re­ anisms underlying the different biological interventions, including NIBS
covery and increased chance of being clinically well compared to and, specifically, tDCS.
collaborative care, (Miklowitz et al., 2007). Yet, a significant proportion Stating the above premises, we conducted two separate systematic
of patients with acute bipolar depression fails to adequately respond to reviews, one dealing with the clinical evidence and the other one with
established pharmacotherapy or psychotherapy, soliciting alternative or the neurobiological underpinnings of the use of tDCS in bipolar
integrative approaches, especially for treatment-resistant cases (Fornaro depressed patients, with the ultimate goal of providing a translational
et al., 2020). perspective on the matter and of highlighting the implications for future
Accruing evidence suggests the potential role in psychiatry of non- research.
invasive brain stimulation (NIBS) approaches, a kind of interventions
underpinned by shared non-invasive and painless delivery of electric 2. Experimental section
current into the brain through the scalp. Their use in patients with
psychiatric disorders is gradually shifting from the research setting to 2.1. Literature search strategy for clinical studies
the clinical practice, resulting also in ad hoc devoted psychiatric neu­
romodulation units (Sauvaget et al., 2018). Among the emerging brain Aiming at achieving a high standard of reporting, we conducted the
stimulation techniques, Transcranial Direct Current Stimulation (tDCS) systematic review of clinical studies owing to the recommendations of
already showed promising results in treating psychiatric conditions such the Cochrane group (Higgins et al., 2016) and PRISMA guidelines (Page
as unipolar depression (Brunoni et al., 2016), auditory hallucinations et al., 2021).
(Mondino et al., 2015) and negative symptoms of schizophrenia (Palm
et al., 2016), obsessive-compulsive disorder (D'Urso et al., 2016), post- 2.1.1. Eligibility
traumatic stress disorder (D'Urso et al., 2018), autism spectrum disor­ Inclusion criteria were: (1) English language studies published in
ders (D'Urso et al., 2015; D'Urso et al., 2014), substance use disorders peer-reviewed journals or records otherwise indexed in peer-reviewed
(Martinotti et al., 2019) as well as various neurological disorders (Fer­ journals, (2) Randomized controlled trials (RCT) comparing active
rucci et al., 2018; Flöel, 2014; Kuo et al., 2014). This technique shows stimulation to sham or to treatment as usual, uncontrolled open-label
great promise also for the treatment of bipolar depression, firstly for its trials, and case reports including patients with an established clinical
safety profile, with only nonspecific and mild adverse effects (i.e. skin diagnosis of BD-I, BD-II, BD not otherwise specified (NOS), not else­
erythema, tingling/itching of the scalp, mild headache, sleepiness), as where classified (NEC) subtypes (depending on the coding system or
well as very low risk of affective switches, rapid cycling or increased edition), confirmed by a semi-structured interview, (3) studies using
suicidality (Bikson et al., 2016). Not less important, it is cheap and tDCS as a tool to specifically treat depression symptoms (e.g., studies
possibly cost-effective compared to the current standard of care (Sau­ using tDCS to treat OCD symptoms in depressed patients or as a primer
vaget et al., 2019). for other treatment approaches were not included), (4) studies providing
TDCS treatment consists in a weak (0.5–2 mA) direct electrical cur­ clinical assessments with depression rating scales before and after tDCS.
rent delivered through one or more sponge electrodes placed on the Exclusion criteria were: (1) studies without clear-cut data about
scalp overlying the targeted cortical areas (Nitsche et al., 2008). Electric patients with bipolar depression, (2) studies with insufficient informa­
current enhances the cortical excitability under the positive electrode tion on mood symptoms (i.e., no depression rating scale scores),
(anode), while an opposite (inhibitory) effect is obtained under the
negative electrode (cathode)(Nitsche and Paulus, 2000, 2001). Other 2.1.2. Search procedure
factors than electrode polarity can influence the tDCS-induced excit­ A systematic search of PubMed was performed with no publication
ability changes, i.e., current intensity and direction (radial vs. tangential date restrictions on October 16th, 2021 using the following search string:
to the cortex surface), session duration, concomitant pharmacological ("mood disorders" OR "bipolar disorder" OR "bipolar") AND ("depression"
treatment, and baseline brain activity (Rahman et al., 2013). In addition, OR "depressive") AND ("tDCS" OR "transcranial direct current
tDCS modulates the long-term effects of use-dependent plasticity, a stimulation").
phenomenon that is believed to be mediated by strengthening synapses Reference lists of identified papers were also screened for additional

2
G. D'Urso et al. Progress in Neuropsychopharmacology & Biological Psychiatry 121 (2023) 110672

studies. The “similar articles” function in PubMed was employed. The Supplementary Text for more details and the combination of terms).
search returned 92 studies (the last interrogation was conducted on
August 8th, 2022). Two reviewers (GDU, ET) independently screened the 2.2.1. Elegibility
title, abstract, and keywords of each study identified by the search and We included publications in peer-reviewed journals and written in
applied the inclusion and exclusion criteria. Discrepancies between the English i) reporting original data or reviewing the molecular and bio­
reviewers, blind to each other, were resolved by consulting a third logical effects of tDCS in bipolar depression; and ii) evaluating the pu­
reviewer (AdB). An additional collaborator with considerable expertise tative mechanisms of action underlying tDCS efficacy in vitro or in vivo,
in the PRISMA methodology and bipolar disorder clinical evidence both in animal models of bipolar disorder or humans suffering from this
(MF), assisted in the critical appraisal of the a priori-defined review condition. Clinical papers not providing an explanatory model of tDCS
protocol. The details of the selection process are summarized in Fig. 1. molecular action were excluded.
The studies included in the qualitative synthesis were 12.
2.2.2. Selection strategy
2.1.3. Data extraction Retrieved records and full-texts were managed by using Endnote X.
Two reviewers independently extracted the following data, when­ Each record retrieved was considered for the coherence of the subject
ever available, according to a-priori-defined abstraction form: (1) de­ with the content of the review by two independent co-authors (FI and
mographic data (sample size, age, sex, concomitant treatment AB). If the abstract text was coherent with the review, the full-text was
strategies); (2) clinical features (diagnosis, active mood symptoms, considered and the reference list was checked to find additional studies.
depression rating scale scores at baseline and study endpoint); (3) tDCS Inconsistencies were resolved by consensus in a meeting with another
stimulation protocol (location of the anode and the cathode, electrode researcher (AdB).
size, current intensity, current density, number and frequency of ses­ The database search and cross-referencing yielded 715 records.
sions, period of stimulation, endpoint data). The study outcome was Finally, 19 articles were included in the qualitative synthesis. The details
based on the scores of validated rating scales administered before tDCS, of the selection strategy are reported in the Prisma 2020 flow diagram
after the acute phase of stimulation, and at subsequent endpoints. (please see Fig. 2).

3. Results
2.2. Literature search strategy for neurobiological studies
3.1. Clinical studies
A comprehensive search in the PubMed was performed on August,
th
8 2022 by using a string combining “tDCS” with 33 terms related to
Fourteen studies were eligible for systematic review (Baliga et al.,
neurotransmitters, signalling molecules, intracellular effectors or targets
2020; Brunoni et al., 2011a; Brunoni et al., 2013a; Gálvez et al., 2011; Li
relevant for the pathophysiology of the bipolar depression (see the

Fig. 1. The PRISMA Flow diagram depicts the flow of information through the different phases of the systematic review.

3
G. D'Urso et al. Progress in Neuropsychopharmacology & Biological Psychiatry 121 (2023) 110672

Fig. 2. The PRISMA Flow diagram depicting the flow of information through the different phases of the systematic review of neurobiological studies.

et al., 2019; Lin et al., 2021; Loo et al., 2012; Loo et al., 2018; Mardani the DLPFC, i.e., the omnilateral electrode system (OLE), optimized for
et al., 2021; Martin et al., 2013; Martin et al., 2011; Palm et al., 2012; peak electric current densities over the DLPFC.
Pereira Junior Bde et al., 2015; Sampaio-Junior et al., 2018) including in All included studies used tDCS as an add-on to medication regimens
total 207 patients with bipolar depression. Data were obtained from the with no drug adjustments during the tDCS administration, except for
publication itself (Baliga et al., 2020; Brunoni et al., 2011a, 2011b; cases of mood switching (Loo et al., 2012; Gálvez et al., 2011). All other
Gálvez et al., 2011; Lin et al., 2021; Loo et al., 2018; Mardani et al., features of the tDCS protocols used in the included studies are reported
2021; Pereira Junior Bde et al., 2015; Sampaio-Junior et al., 2018), by in table 1.
directly contacting the authors (when data from patients with unipolar
depression and bipolar depression were combined) (Li et al., 2019) or 3.2.1. Effects on depressive symptoms
from a previously published review (Dondé et al., 2017) reporting data All the studies included in this review reported reductions of the
obtained by direct contact with the authors of studies with missing in­ depressive symptoms scores over the tDCS treatment. The scales used to
formation (Brunoni et al., 2013a; Loo et al., 2012; Martin et al., 2013; assess the effects on depression symptoms were the Beck Depression
Martin et al., 2011; Palm et al., 2012). The main features of these tDCS Inventory (BDI) (Beck et al., 1996) for four studies (Brunoni et al.,
studies are characterized in Table 1. 2013a; Brunoni et al., 2011a, 2011b; Li et al., 2019; Palm et al., 2012),
the MADRS (Montgomery and Asberg, 1979) for eight studies (Gálvez
et al., 2011; Lin et al., 2021; Loo et al., 2012; Loo et al., 2018; Martin
3.2. tDCS protocols et al., 2013; Martin et al., 2011; Pereira Junior Bde et al., 2015; Sam­
paio-Junior et al., 2018), the Hamilton Depression Rating Scale (HDRS)
In RCTs, sham stimulation refers to the use of the same tDCS montage (Hamilton, 1960) for five studies (Baliga et al., 2020; Brunoni et al.,
used for active stimulation with a ramp of electrical current lasting for 2011a, 2011b; Lin et al., 2021; Mardani et al., 2021; Sampaio-Junior
several seconds at the beginning and/or at the end of 20-min (Loo et al., et al., 2018), and the Quick Inventory of Depressive Symptomatology
2012; Palm et al., 2012) or 30-min sessions (Loo et al., 2012; Sampaio- self-report (QIDS-SR) (Rush et al., 2003) for one study (Loo et al., 2018).
Junior et al., 2018), to elicit weak scalp sensation, while not producing Four studies used more than one scale (MADRS and HDRS in Sampaio-
enduring changes in cortical excitability. Instead, in the 2018 study of Junior et al., 2018; MADRS and QIDS-SR in Loo et al., 2018; BDI and
Loo and colleagues (Loo et al., 2018), the sham consisted of 3 one- MADRS in Li et al., 2019; BDI anf HDRS in Brunoni et al., 2011a, 2011b).
minute ramps delivered 10 minutes apart and a constant current of The reported clinical improvements range from 19 (Brunoni et al.,
0.034 mA throughout the entire session. Sham tDCS data were made 2011a, 2011b) to 31 % (Brunoni et al., 2013a) in the BDI, from 22 (Loo
available from the authors of only two of these studies (Loo et al., 2018; et al., 2018) to 73% (Martin et al., 2013) in the MADRS, and from 18
Sampaio-Junior et al., 2018). (Brunoni et al., 2011a, 2011b) to 92% (Sampaio-Junior et al., 2018) in
While most studies used the 10-20 EEG system for the electrode the HDRS. For the Sampaio-Junior et al. (2018) and the Loo et al. (2018)
positioning, Sampaio-Junior et al., 2018 used a novel tool for targeting

4
Table 1
tDCS studies involving patients with bipolar depression. RCT: Randomized Controlled Study; OPN: Open Label Study; OBS: Observational Study; F3: 10-20 EEG system point for the left dorsolateral prefrontal cortex; F4:
10-20 EEG system point for the right dorsolateral prefrontal cortex; OLE: omnilateral electrode system; Fp2: 10-20 EEG system point for the right superior orbital cortex; F8: 10-20 EEG system point for the right lateral

G. D'Urso et al.
orbital cortex; EC: extracephalic (right upper arm); A: anode; C: cathode; NA: not available; BDI: Beck Depression Inventory; MADRS: Montgomery-Åsberg Depression Rating Scale; HDRS: Hamilton Depression Rating
Scale; QIDS-SR: Quick Inventory of Depressive Symptomatology self-report; SD: Standard Deviation.
Authors, year Study Bipolar Anode Cathode Electrodes Current Current Session N. of Frequency End-point Rating Baseline Endpoint Effect %
Design Patients position position Size Intensity density (A/ duration sessions (weeks) Scales score score Size change
Sample (cm2) (mA) m2) (min) (mean (mean (Cohen’s
+/- SD) +/- SD) d)

Mardani et al., 21.40 ± 1.80 ±


RCTa,b 30 F3 F4 NA 2 NA 20 20 2x/10 days 1.5 HDRS 3.30 92
2021 8.10 2.21
28.7 ±
left- right- 1x/weekday+ 12.0 ±
2 MADRS 5.3 2.66 58
30 DLPFC DLPFC 25 0.80 30 12 Fortnightly 6 7.1
(active) HDRS 23.1 ± 3.39 60
Sampaio- (OLE) (OLE) (10+2) 9.2 ± 4.3
3.9
Junior RCTb
27.9 ± 17.5 ±
et al., 2018c left right- 2 x 0.5 1x/weekday+
MADRS 5.0 9.8 1.34 37
29 DLPFC DLPFC 25 min 0.80 30 12 Fortnightly 6
HDRS 23.5 ± 15.2 ± 1.36 35
(OLE) (OLE) (sham) (10+2)
4.7 7.2
31.05 ± 24.28 ±
MADRS
2.5 5.50 8.96 0.91 22
17 F3 F8 35 0,71 30 20 1x/weekday 4 QIDS-
(active) 15.42 ± 11.17 ± 0.73 28
SR
Loo et al., 4.83 6.70
RCT
2018 30.82 ± 20.00 ±
MADRS
0.034 4.53 8.75 1.55 35
19 F3 F8 35 0,09 30 20 1x/weekday 4 QIDS-
(sham) 15.65 ± 10.00 ± 1.00 36
SR
5.33 5.95
Loo et al., 2 35.5 ± 20.5 ±
RCT 4 F3 F8 35 0,57 20 30 1x/weekday 6 MADRS 2.30 42
2012 (active) 6.5 6.5
Palm et al., 24.0 ± 19.0 ±
RCTd 2 F3 Fp2 35 2/1e 0.57/0.28 20 10 1x/weekday 2 BDI 1.58 21
2012 4.0 2.0
Lin et al., 27.64 ± 17.23
5

OPN 22 F3 F4 35 2 0.57 20 10 2x/5 days 4 HDRS 1.14 38


2021 8.09 ±10.04
1x/weekday+
BDI
Li et al., 2019 OPN 5 F3 F4 35 2 0.57 30 12 Fortnightly 8 NA NA NA NA
MADRS
(10+2)

Progress in Neuropsychopharmacology & Biological Psychiatry 121 (2023) 110672


Pereira Junior
32.8 ± 22 ±
Bde et al., OPN 5 F3 F4 25 2 0.80 30 10 1x/weekday 2 MADRS 1.20 33
2.68 12.4
2015
10.5 ±
Martin et al., 3 MADRS 3.5 8.70 73
OPN 2 F3 F8 35 2 0,57 20 15 1x/weekday 39 ± 3
2013 12 MADRS 16.5 ± 4.44 58
6.5
35 0.57
Martin et al., 2 F3 F8 2 20 15 1x/weekday 3 MADRS 39 ± 3 17 ± 6 4.63 56
OPN A: 35 A: 0.57
2011 1 F3 EC 2 20 20 1x/weekday 4 MADRS 29 15 NA 48
C: 100 C: 0.2
34.21 ± 27.79 ±
Brunoni et al.,
BDI 8.33 11.07 0.65 19
2011a, OPN 14 F3 F4 35 2 0.57 20 10 2x/weekday 1
HDRS 30.77 ± 25.23 ± 0.57 18
2011b
8.96 10.43
Brunoni et al.,
23.21 ± 15.95 ±
2013a, OBS 19 F3 F4 35 2 0.57 20 10 2x/weekday 1 BDI 0.68 31
11.81 9.21
2013b
Baliga et al., Case F3 F4 35 2 0,57 30 10 1x/day 6 HDRS 20 4 NA 80
1
2020 Report F3 F4 35 2 0,57 30 10 1x/day 6 HDRS 25 4 NA 84
Gálvez et al., Case F3 F8 NA 2 NA 20 30 1x/weekday 6 MADRS 42 14 NA 67
1
2011 Report F3 EC NA 2 NA 20 14f 1x/weekday NA MADRS 28 NA NA NA
a
the comparison was not between active and sham tDCS but between medication+active tDCS and medication alone.
b
Enrolling only bipolar depressed patients.
c
Study clearly showing a greater improvement with active versus sham tDCS.
d
Crossover study.
e
1 patient had 1 mA and 1 patient had 2 mA.
f
Treatment interrupted for affective switch.
G. D'Urso et al. Progress in Neuropsychopharmacology & Biological Psychiatry 121 (2023) 110672

studies there is also available information to directly compare the effects now recognized as active elements in the brain and neglected players in
of active versus sham tDCS, with only the former clearly showing a mediating tDCS effects. Monai and colleagues observed a transient
significant superiority of real stimulation, while the second raised astrocytic calcium raise in the cortex of mice treated with tDCS, prob­
serious concerns about the possible biological activity of the sham ably via Ca2+/inositol trisphosphate (IP3) pathway (Monai and Hirase,
protocol used. 2018; Monai et al., 2016) (Fig. 3). In line with these findings, Callai et al.
reported an increase in S100B, a marker of astrocytic activation, in ce­
3.2.2. Safety and tolerance rebrospinal fluid (CSF) of naïve rats after a single session of tDCS (Callai
In the eligible studies, patients with unipolar or bipolar depression et al., 2022). As the Authors used a bimodal model of tDCS, these results
were not considered separately concerning non-specific adverse effects. can be ascribed not only to the active electrode (cathode) which acti­
In particular, six studies (Baliga et al., 2020; Gálvez et al., 2011; Loo vates the cortex, but also to the influence of the reference electrode
et al., 2012; Martin et al., 2013; Palm et al., 2012; Sampaio-Junior et al., (anode) which inhibits cortical regions. Therefore, the behavioural and
2018) reported transient side effects, such as skin redness, itching and biochemical changes observed could depend as much on the activation
tingling at the site of the electrodes, headache, nausea, fatigue, and vi­ as on the inhibition of cortical regions under both electrodes.
sual effects, ranging from mild to moderate intensity. Only one of the Since astrocytic activation may contribute, up to a certain threshold,
examined studies (Loo et al., 2018) reported serious adverse events to many beneficial aspects such as isolation and sequestration of brain
leading to hospital admission: increased suicidality (active treatment, 4 lesions and neuroprotection, tDCS-induced astrocyte activation has
patients), headache and blurred vision related to medication-induced recently been proposed as a relevant molecular target to treat mood
hyponatremia (active treatment, 1 patient), gastroenteritis (sham disorders (Monai and Hirase, 2018), helping to preserve a supportive
treatment, 1 patient), gangrene (active treatment, 1 patient) and stroke environment for the synaptic events of neighbouring neurons (Callai
(active treatment, 1 patient). Nevertheless, all these serious adverse et al., 2022; Monai and Hirase, 2018).
events were assessed as unrelated to study procedures. Solely the case of Mishima et al. found that tDCS affects morphology and motility of
increased suicidal ideation was assessed as possibly related to the tDCS microglia, reducing the surveillance area (Mishima et al., 2019). Of in­
intervention, although it was noted that these patient already had a terest, an increase in serum Glial cell line-derived neurotrophic factor
history of suicidal thoughts and behaviors. (GDNF), a survival promoting molecule which has been firstly charac­
Four cases of an affective switch from depression to hypomania/ terized in striatal astrocytes, has been observed in bipolar patients with
mania were observed with active tDCS (Gálvez et al., 2011; Loo et al., manic symptoms randomized to active tDCS (Lin et al., 1993; Veena­
2012; Martin et al., 2011; Pereira Junior Bde et al., 2015). Furthermore, kumari et al., 2022). Since is well known GDNF ability to promote
Sampaio and colleagues (Sampaio-Junior et al., 2018) described nine dopamine cell survival and differentiation in embryonic midbrain cul­
cases of a treatment-emergent affective switch (five in the sham and four tures (Lin et al., 1993), clinical improvements obtained with tDCS may
in the active group), which did not meet the criteria for a major be mediated by GDNF release and its subsequent effects in the homeo­
depressive episode with mixed features, hypomania, or mania according stasis and maintenance of dopaminergic pathways, thus representing a
to the DSM-5 guidelines and did not require hospitalization, trial protective mechanism to restrain neuronal loss (Fig. 3).
discontinuation or specific treatment. Moreover, a single tDCS session has been shown to reduce inflam­
During tDCS, patients received concomitant treatment with a mood matory mediators levels in the cortex of naïve rats (Callai et al., 2022).
stabilizer and/or antidepressant, which were specified by authors, These results from a preclinical research setting, were paralleled, at a
except Loo and colleagues (Loo et al., 2012; Loo et al., 2018). Neither clinical level, by a 6-week randomized clinical study enrolling patients
rapid cycling induction nor suicidality onset was reported across all suffering from type I or II BD, experiencing a moderate-severe depres­
patients and all tDCS procedures of the examined studies. sive episode. In particular, plasma levels of interleukin (IL)-8 decreased
significantly in the active group compared with the sham-tDCS, and
3.3. Neurobiological studies higher baseline IL-6 levels predicted greater depression improvements
in the active group (Goerigk et al., 2021). Therefore, innate immunity
Targeted stimulation of discrete neuronal networks, enhancing focal biomarkers could be highly sensitive to tDCS delivery. To date, there are
excitability and synaptic strength of learning-related neuronal connec­ no similar studies conducted on bipolar depression patients. However, a
tions (Ghanavati et al., 2022), accompanied by the inhibition of the previous controlled trial enrolling subjects affected by unipolar
excitability of others, would represent a promising way to improve depression, showed that plasma levels of IL-2, IL-2, IL-4, IL-6, IL-10, IL-
neuropsychological deficits characteristic of mood disorders. The 17a, and interferon-γ decreased over time in both the intervention and
neurobiological literature aimed at dissecting the molecular effects of placebo group (Brunoni et al., 2014), raising concerns about the speci­
tDCS is distributed in the mutually dependent and complementary fields ficity of the anti-inflammatory effect of the tDCS. Therefore, although
of synaptic plasticity, neurotransmission, and brain networks (please see these studies are conducted in patients with different mood episodes and
Table 2). results should be interpreted with caution, tDCS response in depression
may depend, at least in part, on its anti-inflammatory action. In light of
3.3.1. tDCS effects on astrocytic homeostasis and neuronal environment the neuroinflammatory model of bipolar depression, supported by
Molecular and neurochemical changes induced by tDCS are still growing evidence encompassing elevated levels of peripheral inflam­
poorly characterized and may affect neuronal survival and plasticity by matory mediators (Grewal et al., 2022), stable immunological alter­
involving multiple biological pathways. ations (Rege and Hodgkinson, 2013; Rosenblat and McIntyre, 2017),
For instance, evidence from in vivo preclinical studies suggests that comorbid systemic autoimmune diseases (Bachen et al., 2009; Chen
tDCS can not only directly stimulate neurons (Takano et al., 2011), but et al., 2021; Farhi et al., 2016; Hsu et al., 2014; Kupka et al., 2002), and
also indirectly stimulate glial cells. It has been proposed that tDCS may genetic findings (Tylee et al., 2018), the anti-inflammatory properties of
influence the astrocyte activation by increasing astrocytic calcium sig­ tDCS may be quite attractive. As inflammatory abnormalities may affect
nalling, significantly influencing the tripartite synapse architecture and the synapse and related transmission (Aguilar-Valles et al., 2020) (Corsi-
functioning. In fact, astrocytes are glial cells filling the space between Zuelli and Deakin, 2021), a targeted anti-inflammatory action on spe­
the presynaptic bouton and the post-synaptic spine, providing a func­ cific brain regions may reverberate on discrete neurotransmitter systems
tional synaptic and vascular interface capable of organizing active and functional circuits.
synaptic connections and supply of nutrients and biochemicals to neu­ How tDCS may impact astrocytic homeostasis, inflammatory path­
rons (Hillen et al., 2018; Monai and Hirase, 2018). In addition to being ways, as well as synaptic plasticity functions remains do be determined,
essential as “nurse” cells supporting neuronal function, astrocytes are but beyond acting on neuronal polarization, it should be noted that tDCS

6
G. D'Urso et al. Progress in Neuropsychopharmacology & Biological Psychiatry 121 (2023) 110672

Table 2
Studies investigating tDCS effects on synapse and transmission.
Authors Model Intervention Molecular effects Functional implications

Monai et al., Transgenic mice in which astrocytes Active anodal tDCS (0.1 mA Active anodal tDCS invoked large-amplitude Anodal tDCS induces astrocytic
2016 and cortical neurons express G- for 10 min) or sham over the and synchronized Ca2+ surges in astrocytes activation through the involvement of
CaMP7, a green fluorescent protein visual cortex across the entire cortex. These enhancements Ca2+ /IP3 signalling. These alterations
Ca2+ indicator; IP3R2 knockout were dependent on the α1 adrenergic receptor may depend on adrenergic receptors.
mice and were not observed in IP3R2 knockout mice
Callai et al., Winstar rats Single session of active Active bimodal tDCS increased S100B levels in Bimodal tDCS may exert an analgesic/
2022 bimodal tDCS (0.5 mA, 20 CSF, decreased pain sensitivity, and cerebral anti-inflammatory effect and induce
min) or sham over the TNF-α levels astrocytic activation
primary cortex
Mishima C57BL/6J and IP3R2 knock-out Anodal tDCS (0.1 mA, 10 Active anodal tDCS altered Iba1 Active anodal tDCS affects the
et al., 2019 mice; BAC-GLT1-G-CaMP7 line 817 min) or sham over the immunohistochemical patterns, enlarged morphology and dynamics of
mice; Iba1-GFP mice primary visual cortex in mice microglial somata, and reduced motility in microglia. These alterations may
under anesthesia or in awake awake mice. depend on adrenergic receptors
conditions
Goerigk et al., Patients with type I or II bipolar 12 sessions of bifrontal tDCS Active tDCS reduced IL-8 plasma levels. tDCS may exert an anti-inflammatory
2021 disorder in a moderate-to-severe (2 mA, 30 min) or sham over Higher levels of IL-6 levels predicted action
depressive episode (n=52) 6 weeks depression improvement in the active group.
Fonteneau Healthy subjects (n=32) Single session of bifrontal Active tDCS induced a significant decrease in tDCS induces striatal dopamine release
et al., 2018 tDCS (20 min, 2 mA) or sham [11C]-raclopride binding in the striatum
Bunai et al., Healthy subjects (n=17) Bifrontal tDCS (2mA, 13 min Active tDCS induced an elevation in GABA tDCS induces a significant increase in
2021 twice at an interval of 20 content in the left striatum. dopamine release in the right striatum
min) or sham tDCS also induced a reduction in [11C]- and GABA concentrations in the left
raclopride binding in the right striatum striatum.
Fukai et al., Healthy subjects (n=20) Bifrontal tDCS (2 mA, 13 min Active tDCS induces a reduction in [11C]- tDCS induces a significant release of a
2019 twice at an interval of 20 raclopride binding in the right ventral striatum dopamine in the right ventral striatum
min) or sham
Meyer et al., Healthy subjects (n=40) Single session of anodal Anodal F4 tDCS increase amplitude of low- tDCS increases spontaneous
2019 prefrontal tDCS (15 min at 2 frequency fluctuations as detected by fMRI mesostriatal neural activity
mA intensity) or inverse
montage
Tanaka et al., Sprague Dawley rats (n=25) Cathodal, anodal, tDCS (800 Extracellular dopamine levels increased for Cathodal tDCS induces an increase in
2013 μA, 10 min) or sham more than 400 min in the striatum as measured striatal extracellular dopamine levels
by in vivo microdialysis after cathodal tDCS.
No significant changes in extracellular
serotonin levels.
Leffa et al., Wistar Kyoto rats (n= 20) 8 sessions of frontal anodal Dopamine levels increased in the hippocampus tDCS significantly increases dopamine
2016 Spontaneous hypertensive rats tDCS (0.5 mA, 20 min) or and striatum in both strains. levels in striatum and hippocampus;
(n=28) sham Striatal BDNF levels increased in Wistar rats. dopamine release may, in turn,
increase neuronal activation by
regulating BDNF levels
Takano et al., Sprague–Dawley rats (n=12) Anodal prefrontal tDCS (400 Signal intensities in the frontal cortex and Anodal tDCS induces neuronal
2011 μA, 10 min) or sham nucleus accumbens were significantly activation in the frontal cortex and its
increased after stimulation as revealed by fMRI connected brain region, especially in
the nucleus accumbens
Hunter et al., Healthy subjects (n=11) anodal tDCS over right After tDCS an increase in Glx under the anodal The excitatory after-effects of anodal
2015 parietal cortex (2.0 mA, 30 electrode was detected by 1H-MRS. As revealed tDCS increases the glutamatergic
min) by fMRI, tDCS increased the within-network signaling and network connectivity
connectivity within the superior parietal,
inferior parietal, left frontal-parietal, salience
and cerebellar intrinsic networks tDCS
decreased the connectivity in the anterior
cingulate and the basal ganglia.
Mezger et al., Healthy subjects (n=20) bifrontal tDCS or sham (2 MRS failed to reveal significant changes of Glu, tDCS led to a reduction of Glu levels in
2021 mA, 20 min) Glx and GABA levels; subgroup analysis the MRS voxel close to the cathode in
revealed a Glu reduction only in female female but not in male participants
participants
Filmer et al., Healthy subjects (n=47) 3 sessions of anodal, cathodal As detected by MRS, subjects who showed a GABA and glutamate concentrations
2019 tDCS (0.7 mA, 9 min), or higher level of inhibition (more GABA relative were linked to the degree of efficacy
sham at left PFC to glutamate) in the prefrontal cortex were
affected by cathodal stimulation to a greater
extent
Alvarez- Healthy subjects (n=23) Bifrontal tDCS (2mA, 20 min) Glx concentrations increased from pre- to post- tDCS over the prefrontal cortex elicits
Alvarado or sham in combination with intervention in the active versus sham group. sustained increase in excitatory
et al., 2021 cognitive training for 2 weeks No difference in GABA concentration was neurotransmitter concentrations
detected by MRS
Rohan et al., Sprague Dawley Rats (n=34) Anodal tDCS (0.10 or 0.25 Ex vivo preparations of hippocampal slices tDCS induces a robust enhancement in
2015 mA for 30 min followed by 30 from rats showed a robust twofold synaptic plasticity
min of recovery time) enhancement in LTP induction and a 30%
increase in PPF
Yu et al., Sprague-Dawley rats (n=224) Active anodal tDCS (250 μA/ Anodal tDCS enhances hippocampal CA1 LTP; Anodal tDCS enhances LTP through
2019 0.25 cm2, 30 min) or sham BDNF protein levels increased in the activation of BDNF/TrkB signaling
over the hippocampus hippocampal CA1 tissue of tDCS-treated rats pathway
compared to sham-treated rats; TrkB
(continued on next page)

7
G. D'Urso et al. Progress in Neuropsychopharmacology & Biological Psychiatry 121 (2023) 110672

Table 2 (continued )
Authors Model Intervention Molecular effects Functional implications

antagonist blocked tDCS-induced


enhancement of LTP ex vivo
Martin et al., Patients with unipolar and bipolar Active anodal tDCS (2.5 mA) Genotype differences in the BDNF Lack of association between BDNF and
2018a, depression, currently experiencing or sham (34 μA) over the Val66Metand COMT Val158Met COMT polymorphisms and the
2018b a major depressive episode (n=120) DLPFC for 30 min each polymorphisms were not associated with antidepressant effect of tDCS.
session for 20 sessions antidepressant effects of active tDCS
Kim et al., Sprague-Dawley rats (n=19) Anodal active tDCS or sham Unilateral anodal-tDCS resulted in significant Anodal tDCS enhance the expression of
2017 (250 μA, 20 min) over the increases in transcription of BDNF, CREB), plasticity-associated genes
right sensorimotor cortex synapsin I, CaMKII, arc in the ipsilateral cortex,
(250 μA, 20 min) for 7 as well as a significant increase in c-Fos and Arc
consecutive days mRNA in the ipsilateral hippocampus

TDCS= transcranial direct stimulation; PFC= prefrontal cortex; PPF= paired-pulse facilitation; LTP= long-term potentiation; IP3R2= inositol trisphosphate receptor
type 2; GFP= green fluorescent protein; CSF= cerebrospinal fluid; IL= interleukin; TNF-α= tumor necrosis factor α; BDNF= brain-derived neurotrophic factor; fMRI=
functional magnetic resosonance imaging; MRS= magnetic resonance spectroscopy; CREB= cAMP response element-binding protein;, CaMKII= Ca2+/calmodulin-
dependent protein kinase II; GABA= γ-aminobutyric acid; COMT= catechol-o-methyltransferase

Fig. 3. The effects of tDCS on the tripartite synapse. tDCS focally increases blood flow and blood-brain-barrier permeability. The stimulation leads to a transient
increase in astrocytic calcium signalling, subsequent astrocyte activation and release of S100B and GDNF. GDNF, in turn, provides support to synaptic plas­
ticity processes.

can increase regional blood flow (Cancel et al., 2018), nitric oxide levels, days elicited dopamine release in the striatum and hippocampus of both
(Barbati et al., 2020) blood-brain-barrier permeability (Shin et al., Wistar Kyoto Rats (WKR) and spontaneous hypertensive rats (SHR)
2020), changing the spatio-temporal solute distribution profile in the (Leffa et al., 2016).
extracellular space (Xia et al., 2020), leading to a widespread series of Other findings support the involvement of dopamine function in the
molecular modifications in the CNS, which ultimately converge on the tDCS mechanism of action. In a double-blind sham-controlled study,
synapse. healthy subjects undergoing a single session of tDCS showed after the
stimulation period a dopamine increase in the ventral striatum
3.3.2. Dopamine neurotransmission compared to sham controls (Fonteneau et al., 2018). An increase in
In vivo studies have convincingly demonstrated a major role for striatal dopamine release was also demonstrated in healthy subjects
dopamine release, dopamine D2/D3 receptors, and dopamine trans­ undergoing [11C]-raclopride positron emission tomography (PET) scans,
porter dysfunctions in bipolar disorders (Ashok et al., 2017; Pearlson after active tDCS delivery but not sham stimulation (Bunai et al., 2021;
et al., 1995; van Enkhuizen et al., 2015; Wong et al., 1997). Multiple Fukai et al., 2019).
studies, both in rodents and in humans, converge in indicating a recip­ Therefore, the increase in dopamine activity and release in subcor­
rocal modulation of dopamine transmission and tDCS effects, suggesting tical areas can secondarily affect the meso-cortico-limbic transmission,
that tDCS impact on dopamine transmission may theoretically be representing a new element to take into account in the mosaic of puta­
beneficial in bipolar disorder (Fonteneau et al., 2018; Ghanavati et al., tive tDCS mechanisms of action, beyond the simple “excitatory-inhibi­
2022; Imburgio et al., 2021; Meyer et al., 2019; Prowacki et al., 2022). tory” model.
Notably, the application of cathodal, but not anodal, tDCS for 10 min It has been observed that the administration of the dopamine pre­
to rodents has been found to increase extracellular dopamine levels for cursor L-dopa may reverse the effects of tDCS on cortical excitability. In
more than 400 min in the striatum. The same protocol of stimulation did a protocol investigating motor cortex stimulation by tDCS in healthy
not change extracellular serotonin levels (Tanaka et al., 2013), thereby subjects, L-dopa abolished the excitability enhancement driven by
implicating a dopamine-specific effect of this tDCS procedure. More­ anodal tDCS while prolonging cathodal tDCS-induced excitability
over, it has been reported that the application of constant electrical of reduction (Kuo et al., 2008). In another study, the effects of increasing L-
0.5 mA intensity on the frontal cortex for 20 min/day for a total of eight dopa doses on motor cortex excitability by tDCS were explored in 12

8
G. D'Urso et al. Progress in Neuropsychopharmacology & Biological Psychiatry 121 (2023) 110672

healthy subjects. The authors found that low and high dosages of L-dopa serotonin facilitates tDCS effects.
abolished both facilitatory and inhibitory potential amplitudes induced
by tDCS, whereas medium dosage prolonged inhibitory potentials, and 3.3.4. Glutamate and GABA neurotransmission
turned facilitatory potentials into inhibition (Monte-Silva et al., 2010b). Glutamate and the GABA/Glutamate balance have become central in
Thus, L-DOPA administration exhibited an inverted U-shaped dose- the arena of antidepressant and mood disorder drug discovery, including
response curve on tDCS-mediated excitability. Similar dose-dependent bipolar depression. In particular, interest in agents acting at glutamate
effects are obtained by administering dopamine agonists such as receptors has grown in the last few years, since these agents could
apomorphine in humans (Fresnoza et al., 2021). represent innovative interventions, particularly for patients who are
A complex interaction between tDCS response and Catechol-Oxy- poor responders to canonical antidepressants or require rapid onset of
MethylTransferase (COMT) genotypes has been reported. In a multi­ the antidepressant action (Tomasetti et al., 2017).
center trial recruiting unipolar and bipolar patients randomized to TDCS has been demonstrated in multiple settings to modulate
receive active or sham tDCS sessions, COMT genotype was not associ­ glutamate and amino acids brain concentration (Santana-Gómez et al.,
ated with the antidepressant effects of tDCS (Martin et al., 2018a). 2015; Alvarez-Alvarado et al., 2021; Hunter et al., 2015).
Another negative result comes from a recent study conducted on healthy As regards the anodal stimulation, a preliminary study sought to
subjects displaying that the COMT Val158Met polymorphism (rs4680) determine the effects of the selective NMDA receptor partial agonist D-
did not impact the beneficial after-effects of prefrontal tDCS on working Cycloserine on the increase of motor cortex excitability in healthy in­
memory (Jongkees et al., 2019). However, other reports indicate a pu­ dividuals (Nitsche et al., 2004a). The authors found that 100mg D-
tative role of COMT genotype in tDCS responses. For instance, in Cycloserine prolonged, but not increased, the anodal tDCS-induced
schizophrenia patients with treatment-resistant auditory hallucinations, excitability enhancement, while no effects were observed on cathodal
the reduction in the auditory hallucination sub-scale score achieved by tDCS-induced excitability reduction (Nitsche et al., 2004a). These data
add-on tDCS was significantly potentiated in subjects carrying the lend support to the idea that the duration of anodal-mediated excit­
COMT Val genotype (Chhabra et al., 2018). Furthermore, the COMT Val ability enhancement may be under the control of glutamatergic mech­
genotype was found to crucially modulate the efficacy of cathodal tDCS anisms. In a more recent study including healthy volunteers, the authors
to the dorsolateral PFC on response inhibition, a core aspect of executive tested whether tDCS effects on cortical excitability were modulated by
function, as highlighted during the performance of a parametric Go/No- the interaction between nicotine and glutamate receptors since both
Go test in healthy volunteers (Nieratschker et al., 2015). affect cortical plasticity via calcium channels. Therefore, anodal tDCS
Taken together, these findings indicate a complex pattern of in­ was combined with 15 mg nicotine patches and three doses of dextro­
teractions between the dopaminergic transmission and tDCS, worthy of methorphan. Nicotine was found to abolish anodal tDCS-induced motor
further investigations that could prove relevant for the treatment of the cortex excitability enhancement, an effect that was prevented by an
bipolar disorder. average dosage of dextromethorphan (Lugon et al., 2017). Low-dose
dextromethorphan did not affect the impact of nicotine on tDCS-
3.3.3. Serotonin neurotransmission induced plasticity, while high-dose dextromethorphan abolishing plas­
Dysfunctions in serotonin neurotransmission have been consistently ticity. On the other hand, administration of the GABA receptor agonist
reported in bipolar disorder (Chou et al., 2010; Gao et al., 2016; lorazepam, that is used as an anxiolytic and tranquilizing agent in many
Oquendo et al., 2007). An in vivo microdialysis study conducted in rats psychiatric diseases including bipolar disorder, resulted in a delayed,
failed to detect any changes in striatal serotonin levels following cath­ but then enhanced and prolonged anodal tDCS-induced excitability
odal or anodal tDCS (Tanaka et al., 2013). Although only a few reports elevation (Nitsche et al., 2004b)
have investigated the potential role of serotonin in mediating the effect Concerning the cathodal stimulation, a randomized magnetic reso­
of tDCS, it has been repeatedly demonstrated that serotonergic agents nance spectroscopy (MRS) and connectivity study on twenty healthy
enhance tDCS-mediated neuroplasticity in humans (Nitsche et al., subjects observed no changes in glutamine and GABA concentration in
2009), thereby putatively implicating serotonin neurotransmission in the stimulated right DLPFC before, during, and after tDCS procedures.
the neurobiology of tDCS effects in bipolar disorder. However, when considering only the data from the twelve female par­
Notably, a single dose of the Selective Serotonin Reuptake inhibitor ticipants, a Glutamate reduction after active compared to sham stimu­
(SSRI) citalopram in healthy subjects has been found to increase both lation was detected. No changes were observed in resting-state
the amplitude and duration of the after-effects of anodal tDCS. Addi­ functional connectivity (Mezger et al., 2021). Furthermore, research
tionally, citalopram counterbalanced the diminution in excitability after targeting the motor cortex revealed that cathodal tDCS is able to reduce
cathodal tDCS, inducing facilitation (Nitsche et al., 2009). Chronic an­ the concentration of Glx in the site of stimulation (Stagg et al., 2009) and
tidepressant treatment with citalopram for 35 days increased and pro­ of GABA in the contralateral cortex (Bachtiar et al., 2018). A different
longed the LTP-like plasticity induced by anodal tDCS for over 24 h, and MRS study showed that a higher baseline GABA/Glutamate ratio in­
converted cathodal tDCS-induced long-term depression (LTD)-like creases the effect of cathodal tDCS over the left DLPFC in disrupting the
plasticity into facilitation. These effects were abolished by the N-methyl- performance in a response selection training (Filmer et al., 2019).
D-aspartate (NMDA) receptor antagonist dextromethorphan, putatively The above findings reinforce the view that tDCS-mediated changes of
indicating that the potentiating effects of SSRI administration on tDCS cortical excitability are under a relevant and multimodal glutamatergic
procedures may be NMDA-receptor dependent, and may be compatible and GABAergic control. However, to date, no specific studies have been
with a neuromodulatory, but not a plasticity-driving function of sero­ conducted in bipolar depression or other mood disorders and therefore
tonin (Kuo et al., 2016). In contrast to what we have previously reported this is an area needing further investigations, especially after the
with L-dopa and dopamine agonists, the effects of serotonergic agents on approval by regulatory agencies of the first glutamatergic drug, esket­
anodal tDCS-induced excitability appear to be independent of the dose amine, for the treatment-resistant depression (Kim et al., 2019; Popova
(Melo et al., 2021). et al., 2019).
The effects of tDCS on the serotonergic system and therefore the
differential sensitivity, as well as the inter-subject variability, could be at 3.3.5. Synaptic plasticity and brain networks putatively involved in the
least in part influenced by specific variants of the serotonin transporter pathophysiology of bipolar depression: relevance for tDCS
gene (5-HTTLPR) (Brunoni et al., 2013b). TDCS has been considered to improve cognitive functions by
In summary, serotonin can reverse the cathodal and enhance the enhancing long-term potentiation (Fig. 4), an effect that could have
anodal effects of tDCS and the evidence suggests a bidirectional inter­ therapeutic implications in unipolar and bipolar depression (McClintock
action mode: tDCS promotes the function of the serotonergic system and et al., 2020). An enhancement of hippocampal CA1 LTP has been shown

9
G. D'Urso et al. Progress in Neuropsychopharmacology & Biological Psychiatry 121 (2023) 110672

Fig. 4. Anodal tDCS increases the expression of immediate early genes such as Arc, c-Fos, BDNF and others, thus enhancing long-term potentiation. BDNF= Brain-
derived neurotrophic factor; AMPAR= α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor; NMDAR= N-methyl-D-aspartate receptor; LTP= long-term
potentiation.

in rodent slices up to 1 week after tDCS (Rohan et al., 2015). Yu et al. synapses constituted by a mesh of receptors (i.e.: NMDAR; α-amino-3-
have demonstrated that, in rats, tDCS-induced LTP is associated also hydroxy-5-methyl-4-isoxazole propionic acid receptor, AMPAR; Gluta­
with memory enhancement in the hippocampal-dependent passive mate Metabotropic Receptor type I, mgluR1 and 5), scaffolding, and
avoidance learning task, which is blocked by the pretreatment with adaptor proteins (i.e. Homer, PSD 95; Shank)(de Bartolomeis et al.,
ANA-12 (Yu et al., 2019). These animals also showed an increase in 2022b; Iasevoli et al., 2013). Moreover, multiple lines of evidence
Brain-Derived Neurotrophic Factor (BDNF) in the CA1 region of hip­ demonstrate the involvement of PSD proteins in the response to treat­
pocampus compared to sham-control animals (Fig. 4). In agreement ments for psychiatric ilnesses including bipolar disorders (Barone et al.,
with these findings, albeit in different experimental conditions, tDCS has 2021; de Bartolomeis et al., 2022a; Halff et al., 2021; Iasevoli et al.,
been found to enhance expression and dendritic spine density in the 2020). Mood stabilizers administration has been found to modulate
peri-infarct motor cortex of mice subjected to focal ischemia of the shaping and composition of postsynaptic structures by affecting the
motor cortex (Longo et al., 2022). Taken together, these results suggest expression of Homers, Shanks, and others PSD proteins (de Bartolomeis
the possibility that tDCS-induced cortical plasticity may be mediated, at et al., 2012; Halff et al., 2021; Tomasetti et al., 2011). A relevant step in
least in part, by BDNF/TrkB pathway. The putative role of BDNF/TrkB in the signaling cascades potentially involved in synaptic plasticity events
the mechanism of tDCS effects is intriguing, given the pathogenic and is represented by the induction or activation of immediate early genes
therapeutic role proposed for this signaling in major depression and (IEGs) (Cirillo et al., 2017; de Bartolomeis et al., 2013; de Bartolomeis
bipolar disorders (Chiou and Huang, 2019; D'Addario et al., 2018; et al., 2022b), which are genomic elements whose expression is rapidly
Schröter et al., 2020; Valvassori et al., 2019), as well as for the supposed upregulated after neuronal activation. IEGs code for transcription fac­
involvement of the BDNF/TrkB pathway in the mechanism of action of tors (i.e., c-fos) or more rarely for molecules implicated in synaptic ul­
lithium (De-Paula et al., 2016). trastructure (i.e., Arc) or intracellular signaling pathways (Gallo et al.,
Once reviewed the evidence on the effect of tDCS on synaptic plas­ 2018). At the biological level, IEGs are predicted to induce rapid and
ticity and its potential implication in bipolar disorders, a further step is transient molecular effects in neurons, which may have a strong impact
to consider if and how the evidence supports an after-effect on synaptic on neuronal plasticity (Sommerlandt et al., 2019). tDCS has been re­
architecture and dendritic spine whose aberrations at both a structural ported to induce multiple early genes (Kim et al., 2017; Ranieri et al.,
and functional level have been described in the molecular pathophysi­ 2012), including c-fos and Arc. In particular, mRNA levels of BDNF,
ology of bipolar disorders (Alfieri et al., 2017; Konopaske et al., 2014; cyclic AMP response element-binding protein (CREB), synapsin I, Arc,
Law et al., 2004; Martin et al., 2018b; Nanou and Catterall, 2018). At the and c-Fos were found significantly increased in the ipsilateral cortex
cellular level, tDCS has been shown to induce changes in dendritic spines after 7 days of unilateral intervention, whereas only c-Fos expression
density and structure (Cirillo et al., 2017). Recently, postsynaptic den­ increased in the ipsilateral hippocampus (Kim et al., 2017)(Fig. 4).
sity (PSD) has emerged as a key structure of the synapse dysfunction in Notably, has also been observed an increase after tDCS of CaMKII gene
psychiatric disorders, including major depression and bipolar disorders expression, a molecule robustly correlated to the mechanism of action of
(Akula et al., 2016; de Bartolomeis et al., 2014a; de Bartolomeis et al., drugs with antidepressant effect (Celano et al., 2003; Tiraboschi et al.,
2014b; de Bartolomeis and Tomasetti, 2012; Föcking et al., 2016). PSD 2004).
is a cytoskeletal structure beneath the plasma membrane of excitatory It is worth mentioning that a different form of NIBS, i.e. transcranial

10
G. D'Urso et al. Progress in Neuropsychopharmacology & Biological Psychiatry 121 (2023) 110672

magnetic stimulation (TMS), has been demonstrated to induce Homer1a in the membrane potential. Moreover, in a pilot study from the same
in the cingulate cortex after swimming stress an animal model of group (Pereira Junior Bde et al., 2015), it was argued that the rate of
depression-like behavior (Sun et al., 2011). Homer1a is both an response and remission following tDCS in bipolar patients were similar
immediate-early gene and a key structural and functional member of to those observed in unipolar ones, i.e. 40% and 20% respectively.
PSD, involved in dendritic spine formation and architecture (de Barto­ Accordingly, a recent open-label trial directly compared the response to
lomeis et al., 2022b; Meyer et al., 2014), implicated in depression tDCS of unipolar and bipolar depressed patients and showed that both
pathophysiology as well as in the response to pharmacological and non- groups improved equally not only in depression scores, but also in
pharmacological treatments (Serchov et al., 2015). Taking the TMS cognitive performance, anxiety, and psychosocial functioning (Lin et al.,
study as a basis, it would be of interest to find out if also tDCS can 2021).
modulate Homer1a and in which brain areas. On the contrary, Li and colleagues reported a higher rate of treat­
It will be interesting for a better understanding of tDCS effects on ment response in bipolar patients (40%) than in unipolar ones (30,8%)
synaptic plasticity and dendritic architecture to test in further studies (Li et al., 2019). Interestingly, in this trial a statistically significant
whether tDCS can also modulate a relevant dendritic spine shaper such reduction of the MADRS score was not present at the end of the treat­
as Homer and its interacting proteins, specifically with the regard to the ment and was observed only four weeks later, pointing to a delayed
potential involvement of dopaminergic system that has been demon­ antidepressant effect of tDCS. Unfortunately, the authors did not specify
strated to have a major effect on postsynaptic gene expression (Toma­ if there was any difference in the timing of response between unipolar
setti et al., 2007) and that has already been suggested to have a and bipolar patients.
significant role in bipolar disorders pathophysiology both for the Brunoni and colleagues (Brunoni et al., 2013a; Brunoni et al., 2011a,
depressive and manic phases of the disease (McCutcheon et al., 2021; 2011b) were the first researcher to suggest that tDCS in bipolar
Salvoro et al., 2018). depression could have at least the same efficacy and tolerability as in
We refer to an earlier publication for a more complete overview of unipolar depression, so extending the claims of the earliest reports
neurobiological mechanisms of tDCS effects (McLaren et al., 2018), describing tDCS as a promising emerging therapy for major depression
which is beyond the scope of the present review that focused specifically (Boggio et al., 2008; Ferrucci et al., 2009). Indeed, the authors observed
on neurobiological mechanisms relevant to bipolar disorder. that in the clinician-rated measures of depression, symptoms improved
exclusively in patients with bipolar depression, although in that study
4. Discussion the total sample was relatively small and the sample of bipolar patients
was smaller than that of unipolar patients (14 bipolar vs. 31 unipolar).
We submit that the present review is prime since it specifically aimed This surprising finding has been interpreted as the result of a difference
at appraising evidence of tDCS efficacy in bipolar depression, a critical in the baseline depression severity between the two groups, with bipolar
area of research, and concurrently discusses its neurobiological un­ patients scoring much higher on clinician-rated scales (30.77 vs. 21.53
derpinnings. Whilst very informative about the clinical effects of tDCS, HDRS score). Moreover, in a meta-analysis from the same group, bipolar
the two systematic reviews on the matter carried by Donde’ and col­ depression has been suggested to be a positive predictor of tDCS
leagues (Dondé et al., 2017; Dondé et al., 2018), could not include the response, even if the number of bipolar patients in the overall sample of
most current results, i.e. two open label studies (Li et al., 2019; Lin et al., the study was as low as 3.8% (Brunoni et al., 2016).
2021) and three RCTs (Loo et al., 2018; Sampaio-Junior et al., 2018; Loo and colleagues (Loo et al., 2012) confirmed the antidepressant
Mardani et al., 2021). In particular, the RCTs by Sampaio and colleagues effects of tDCS on a mixed sample comprising unipolar (N=52) and bi­
(Sampaio-Junior et al., 2018) and by Mardani and colleagues (Mardani polar (N=8) patients, with a significantly greater improvement in the
et al., 2021) are the only specifically designed for bipolar depressed active group compared with the sham group over the 3-week study
patients available to date and, taken together, enrolled more patients period on the primary outcome measure (change in MADRS). The results
with bipolar depression than all the previous studies in aggregate. were clinically modest, with only a 28% decline in MADRS scores after
Moreover, our study reviews the neurobiological literature to dissect the active stimulation and a small proportion of responders in the whole
potential underpinnings of the tDCS effect in bipolar depression, in sample. Of note, the four bipolar patients allocated in the active group
terms of impact on neurotransmitter action, intracellular signaling, showed an average greater improvement, i.e., 42%. In a subsequent
synaptic plasticity, and brain networks modulation. randomized double-blind sham-controlled trial on a mixed sample of
unipolar (N=84) and bipolar (N=36) patients, the same group of authors
4.1. tDCS efficacy for the treatment of bipolar depression suggested that the modest overall mood improvement – with no statis­
tically significant difference between active and sham stimulation – may
Several RCTs and meta-analyses have demonstrated the efficacy of have been caused by a greater placebo effect in the sham group (Loo
tDCS in major depressive episodes (Moffa et al., 2020; Mutz et al., 2019; et al., 2018).
Razza et al., 2021). However, most studies examining tDCS in the This hypothesis stimulates reflection on the different sham stimula­
treatment of depressive symptoms have enrolled both unipolar and bi­ tion protocols currently used in tDCS research and their possibly
polar depression patients, without corresponding stratification, thus different biological effects. Indeed, the inconsistency in sham protocols
hindering the detection of eventual differences in the tDCS effects be­ among sham-controlled trials might be partially responsible for the
tween the two groups. inconsistency of their clinical results (Fonteneau et al., 2019). In the
One of the few studies specifically addressed to bipolar depressed study by Loo et al. (2018), the authors speculated that the special sham
patients, by Sampaio and colleagues (Sampaio-Junior et al., 2018), was stimulation used (consisting in a 0.034 mA of constant current during
a randomized double-blind sham-controlled trial that confirmed the the whole session plus two ramps of 1 minute up to 0,5 or 1 mA) may
efficacy and safety of add-on prefrontal tDCS in a sample of patients with have been biologically active, and, when given for 20 sessions over 4
BD type I or II who were in a major depressive episode. In particular, this weeks as in this study, could have had comparable effects to 1mA and 2
study showed that active tDCS was superior to sham for a sustained mA continuous stimulation, so leading to a non-significant difference
response but not for sustained remission. Importantly, similar rates of between active and sham group at the end of the study.
treatment-emergent affective switches were reported in active and sham The other three RCTs included in this systematic review (Loo et al.,
treatment groups. In their discussion, the authors hypothesized that 2012; Palm et al., 2012; Sampaio-Junior et al., 2018) used a different
greater beneficial effects might be achieved by combining tDCS with sham procedure which entailed an active stimulation of only 30 seconds
other treatments, i.e. pharmacotherapy, psychotherapy, or other brain with a ramp up and a ramp down identical to that used for the active
stimulation techniques, since tDCS alone can induce only small changes tDCS group and with either the same (Palm et al., 2012; Sampaio-Junior

11
G. D'Urso et al. Progress in Neuropsychopharmacology & Biological Psychiatry 121 (2023) 110672

et al., 2018) or half (Loo et al., 2012) the current intensity. In our In summary, despite the paucity of specific studies, the existing ev­
opinion, this last sham procedure is the best option to date, being less idence suggests that tDCS is effective in treating symptoms of bipolar
likely to produce an appreciable biological effect still ensuring the depression and that it is at least as beneficial in this clinical condition as
subject blinding thanks to the skin sensation induced. In fact, in the in unipolar depression, for which it is already a consolidated treatment.
Sampaio-Junior et al. (2018) study, the patients were assessd for the Moreover, there are clues indicating that bipolar depressed patients
blinding and resulted unable to guess their actual group beyond chance. could benefit from tDCS even more than unipolar ones, both in terms of
Palm and his group (Palm et al., 2012) administered prefrontal tDCS acute response and of long-term effects, especially when appropriate
to 20 unipolar and 2 bipolar patients for only two weeks, obtaining no protocols of continuation tDCS are implemented after an acute response.
meaningful therapeutic effect on their treatment-resistant depression
compared with placebo, even though subjective mood ratings using 4.2. Interaction between tDCS and pharmacotherapy
Positive and Negative Affect Scale (PANAS) showed an increase in
positive emotions and a trend towards a reduction of negative emotions Some of the articles included in this review analyzed the effect of
after active tDCS. Authors suggested that this lack of efficacy could be concomitant drugs taken during the tDCS treatment, a particularly
attributed to a higher degree of treatment resistance or a relatively important issue in the case of bipolar patients, who often need chronic
higher mean age in enrolled patients - compared with previous tDCS regimens of psychotropic medications.
studies - as an older age may be associated with a poorer antidepressant In a pioneering study on healthy subjects, Nitsche et al. observed that
response to transcranial brain stimulation. In our opinion, the disap­ carbamazepine abolished the excitability enhancement induced by
pointing results of this trial were due mainly to the low number and anodal stimulationon motor cortex (Nitsche et al., 2003). Subsequently,
duration of tDCS sessions, factors that had not yet demonstrated as the same group of researchers observed that administration of loraze­
crucial at the time of this study. pam to healthy individuals before tDCS led to a delayed, but more
Martin and colleagues (Martin et al., 2013) examined the use of pronounced and prolonged enhancement of the motor cortical excit­
continuation tDCS treatment to prevent relapse following clinical ability induced by the stimulation (Nitsche et al., 2004b). However, the
response to an acute course of tDCS. The sample of their study consisted mechanisms underlying this peculiar effect remain unclear.
mostly of patients who had failed at least one adequate course of anti­ In depressed patients, Brunoni and colleagues (Brunoni et al., 2013a)
depressant treatment before receiving acute stimulation. They observed observed that benzodiazepine use was associated with higher endpoint
that continuation tDCS, given weekly for the first 3 months, resulted in depression scores, probably because the long-term changes in the
the majority of responders remaining well and that higher medication GABA/glutamate system induced by these drugs were responsible for
resistance was predictive of relapse during this treatment period. Again, abolished tDCS effects. Moreover, as baseline depression severity was
these results refer to a mixed sample and it is not possible to clarify the similar for patients using and not using benzodiazepines, their use might
differential effect on unipolar (N=20) and bipolar (N=3) patients. be a proxy for anxious depression and, thus, a predictor of poor tDCS
Another strategy to maintain the clinical effect of tDCS in the long response. As to antidepressants, the authors observed that they generally
term was investigated by Aparicio and colleagues (Aparicio et al., 2019), increased tDCS effects and suggested that chronic use of serotoninergic
who compared the effects of the same maintenance protocol in unipolar drugs might still induce global facilitation for brain activation, so paving
and bipolar depression. Twenty-four patients (16 with unipolar the way for tDCS action. The same group, in two different studies
depression and eight with bipolar depression) who presented acute tDCS (Aparicio et al., 2019; Brunoni et al., 2011a, 2011b), ascribed to phar­
response (≥50% depression improvement in the Hamilton Depression macotherapy the shorter-lasting effects induced by tDCS in patients with
Rating Scale [HDRS] after receiving 15 tDCS sessions) were followed for MDD compared to those with BDD. In particular, mood stabilizers (used
up to 6 months or until relapse, defined as clinical worsening and/or only in patients with bipolar depression), played a role in maintaining
HDRS > 15. During the follow-up, tDCS was administered twice a week the initial tDCS-induced improvement, whereas antidepressants (used in
(maximum of 48 sessions) over 24 weeks with results of a low relapse both groups) did not.
rate (26,5%) after a 6-month follow-up in tDCS responders, particularly Finally, a recent RCT aimed specifically to compare the effect of
in non-treatment-resistant patients. Interestingly, even if at the end of mood stabilizers (Lithium Carbonate+Sodium Valproa­
the study no significant difference in relapse rates was observed between te+Carbamazepine) alone with that of mood stabilizers combined with
unipolar and bipolar patients, these last relapsed on average 3 months tDCS in bipolar depressed patients (Mardani et al., 2021). The results of
later. The authors suggested that this finding could be ascribed to the this trial indicate that depressive symptoms are reduced more in the
fact that bipolar and not unipolar patients were allowed to take mood tDCS+drugs group compared to the drugs-alone group and that in the
stabilizers during the trial and that probably these drugs have contrib­ tDCS+drugs group there was also an improvement in executive func­
uted to the longer survival of the tDCS effect. tions (i.e. response inhibition), which was not observed in the drugs-
The issue of the follow-up efficacy of tDCS in major depressive alone group. These effects were observed immediately after the two-
episode (MDE) has been specifically assessed by a recent systematic week intervention but were not maintained at a three-month follow-up.
review and meta-analysis including 11 studies that followed up adults In summary, studies involving depressed patients suggest that tDCS
patients undergoing and/or not undergoing tDCS maintenance sessions beneficial effects could be reduced by concomitant benzodiazepines,
in the follow-up period (Razza et al., 2021). According to the authors, increased by serotonergic agents and mood stabilizers, and prolonged
the observed reduction of depression scores at follow-up, compared to only by mood stabilizers. However, evidence is still too scarce to allow
the end of the acute tDCS treatment, was due to the contribution of the for any clinical recommendation on how to combine tDCS with drugs.
studies involving maintenance tDCS sessions. Considering only the trials
including bipolar depressed patients and providing maintenance tDCS 4.3. Impact of tDCS protocols on safety and efficacy
(Aparicio et al., 2019; Loo et al., 2012; Loo et al., 2018; Martin et al.,
2013), the review shows that all of them reported a further improvement In the analyzed articles, a few non-specific tDCS side-effects, tran­
at follow-up, with the exception of Loo et al., which reported no change. sient and low in intensity, were reported. They consisted in skin redness
Regarding the protocol used and the duration of the maintenance and itching/burning/tingling at the site of the electrodes, headache,
treatment, the selected studies differ considerably, with one of them nausea, fatigue, visual effects, agitation and were apparently montage-
performing 48 twice-per-week sessions (Aparicio et al., 2019), two and dosage-independent, provided that general safety recommendation
providing only four once-per-week sessions, and another involving a were followed (Bikson et al., 2016). The occurrence of anxiety was also
more complex schedule of 18 sessions, 12 of which performed once reported as a side-effect in depression studies, even though one trial
weekly, and the remaining six once fortnightly. involving only bipolar patients (N=60) seems to point to a specific anti-

12
G. D'Urso et al. Progress in Neuropsychopharmacology & Biological Psychiatry 121 (2023) 110672

anxiety action of the bifrontal tDCS montage in this category of patients However, two of the four participants who had switched from depres­
(Pereira Junior Bde et al., 2015). sion to hypomania/mania in the examined articles had been treated with
Due to the small number of studies, the relative heterogeneity of the bifrontal tDCS stimulation (Loo et al., 2012; Pereira Junior Bde et al.,
protocols and endpoints used, it is hard to conclude about a differential 2015). Thus, we underline that hypomanic/manic switches are also
effectiveness of the various electrodes montage and/or of the other as­ possible with more focused bifrontal montages. Importantly, no study
pects of the tDCS procedure. However, while most studies used the reported an increase in rapid cycling or suicidality.
conventional 10-20 EEG system to position the electrodes over the target Regarding the other stimulation parameters, as the stimulus intensity
areas, the largest sample RCT used the OLE, a novel electrode montage and duration, the number and frequency of sessions, and the overall
specifically developed to achieve an improved targeting of the bilateral dosage of current administered, their optimal values for tDCS thera­
DLPFC (Sampaio-Junior et al., 2018). Intriguingly, at the six-week peutic effects are still unknown. As a possible reference, we know that in
endpoint the authors reported the most marked symptom improve­ rTMS studies longer treatments led to better results (Gross et al., 2007).
ment among the RCTs, if not considering the study by Mardani et al. However, recent tDCS dose-outcome studies in healthy cohorts sug­
(2021), with a much closer endpoint (ten days). gested that there is not a simple linear association and that there may be
Previous computational modeling research demonstrated that the considerable inter-individual variability in response (Chew et al., 2015).
proximity of the two electrodes to one another is correlated to stimulus Loo and colleagues suggested that stimulation at 2.5 mA for 30 min over
focality, with a reduced amount of current reaching the deep brain 20 sessions probably exceeds the optimal stimulus “dosage” for many
structures and an increased amount shunting superficially across the patients, even though they acknowledge that the exact influence of the
scalp (Datta et al., 2008). How this can influence the clinical effect re­ single stimulus parameters are yet unknown (Loo et al., 2012, 2018).
mains to be determined. The optimal repetition interval for tDCS sessions is also a matter of
As a matter of fact, the F3-F8 and F3-F4 montages have been used in debate. Studies of motor cortex excitability report that the duration of
most clinical trials reporting significant antidepressant effects and both tDCS-induced plasticity is related to the stimulation duration and that
are known to induce relatively high electric fields in dorsolateral pre­ the second stimulation should be optimally timed to coincide with the
frontal cortices. However, by increasing the distance between tDCS after-effects of the first, but not when homeostatic brain regulation oc­
electrodes, deeper limbic structures are stimulated, such as the amyg­ curs because that period might be of limited neuroplasticity (Monte-
dala, hippocampus (Sadleir et al., 2010), and corticolimbic networks Silva et al., 2010a). However, these neurophysiological principles are
(Bikson et al., 2008). hard to be translated into treatment protocols. As a matter of fact, the
Martin and colleagues observed that tDCS antidepressant efficacy four clinical trials included in this review that involve twice daily tDCS
improved after moving the cathode from F4 to an extracephalic position, sessions do not specify the exact timing of the second daily session, with
i.e. switching from bifrontal to fronto-extracephalic montage, although three studies reporting only the minimal interval between the sessions, i.
these observations stem from the treatment of a small and mixed sample e., three (Li et al., 2019) or four (Brunoni et al., 2011a, 2011b; Brunoni
(9 unipolar, 2 bipolar) (Martin et al., 2011). A fronto-extracephalic et al., 2013a, 2013b) hours, and another study not mentioning at all this
montage may induce greater antidepressant effects by more direct and aspect (Mardani et al., 2021).
widespread activation of limbic areas associated with the pathophysi­ In summary, the differential impact on unipolar and bipolar
ology of depression, including the anterior cingulate, the insula, the depression of the many variables involved in tDCS protocols is far from
basal ganglia, and the superior temporal gyrus (Bai et al., 2014; Bikson being ascertained. The only constant in the reviewed evidence is the
et al., 2008). Moreover, preliminary results showed that alternative anodal stimulation of the left DLPFC to obtain the antidepressant effect.
tDCS montages, such as bitemporal electrodes positioning, might also be The position of the cathode varies among studies and could influence the
useful for the treatment of depression (Ho et al., 2015). magnitude of the antidepressant effect (the greater the distance between
A potential major advantage of tDCS in bipolar depression is the electrodes, the greater the effect) and the action on other concurrent
considered to be its low manic-switch potential, compared to standard symptoms. As regards to the dosage of current, while it appears that a
antidepressant therapy (Fornaro et al., 2018). The present review is in greater number of tDCS sessions is necessary to obtain better clinical
agreement with this consideration. Indeed, in the eligible studies, four results, there is no indication available on possible differences in the
cases of an affective switch from depression to hypomania/mania were number of sessions needed to treat unipolar or bipolar depression.
reported. In these cases, tDCS was administered using a bifrontal (F3-F4:
Pereira Junior Bde et al., 2015)(F3-F8: Loo et al., 2012) or a fronto- 4.4. Neurobiology of tDCS effect in bipolar depression
extracephalic montage (F3-EC: Martin et al., 2011; Gálvez et al.,
2011). Martin and colleagues emphasized that their participants had not The neurotransmission involvement in tDCS effects on bipolar
previously reported hypomanic episodes with bifrontal tDCS and that depression can be inferred from studies that explored the relationship
the onset of hypomania had occurred with the fronto-extracephalic between tDCS, monoamine release, and reuptake in the preclinical and
montage. Thus, the extracephalic stimulation may be more likely to clinical setting (Kuo et al., 2016). The most relevant information found
induce affective switches from depression to hypomania/mania, in neurobiological studies concerns dopamine, serotonin, glutamate,
through a greater stimulation of limbic regions, cerebellum and its and the reciprocal interaction between these neurotransmitters.
associated networks, involved in the pathophysiology of mania (Brooks Moreover, multiple lines of evidence suggest an impact of tDCS on
et al., 2010; Minichino et al., 2014). Nevertheless, a higher potential of synaptic strength, synaptic plasticity, and metaplasticity (Cirillo et al.,
affective switch for a specific tDCS montage might also be considered a 2017; Yu et al., 2019). These effects of tDCS are specifically pertinent for
proxy for a more robust antidepressant action, a possibility that deserves bipolar disorders whose molecular pathophysiology is conceptualized as
to be investigated through a direct comparison between the different a disorder of synaptic plasticity and neuronal architecture involving
montages in terms of their respective safety and efficacy. Indeed, a tDCS multiple processes (Akula et al., 2016), from receptors recycling and
montage involving the stimulation of the left DLPFC in combination scaffolding proteins re-arrangement to the induction of transcription
with that of a remote, non-frontal area such as the right cerebellum has factors, then leading to the synthesis of key molecules relevant for
yielded intriguing evidence. In particular, it was reported a mood in­ dendritic spine shaping.
crease in healthy subjects (Newstead et al., 2017), an improvement of The results reviewed point to the involvement of multiple neuro­
sleep (Minichino et al., 2014), neuropsychological performance (Bersani transmitter systems, overlapping signaling systems at the level of second
et al., 2016), and neurophysiological parameters (Bersani et al., 2015) in messenger and transcriptional control, and, finally, to significant
euthymic bipolar patients, and a reduction of abnormal behaviors, changes in brain plasticity and metaplasticity involving proteins that are
motor tics and epileptic activity in autistic patients (D'Urso et al., 2021). integral part of the dendritic spine.

13
G. D'Urso et al. Progress in Neuropsychopharmacology & Biological Psychiatry 121 (2023) 110672

It should be aknowledged that it is premature to speculate if the “Retardation” factors of the scale. Similar studies aiming at the identi­
changes at neurobiological level are just an epifenomenon or a change of fication of response predictors specifically in bipolar depression may
significant functional impact in tDCS mechanism of action. However, it have substantive clinical implications. In particular, since the mild an­
is worth underlining that the changes described in animal models where tidepressant response to tDCS may be due to the heterogeneity of
the neurostimulation is used without interference of other therapeutics selected patients, the detection of response predictors could reveal
and in controlled context are in line with changes expected from a pu­ which subgroup of bipolar depressed patients would benefit more from
tative antidepressant effect. the treatment. Finally, despite preliminary indications, the exact nature,
In summary, even if to date no studies have specifically investigated and the potential synergistic action between tDCS and specific
the neurobiological effects of tDCS in bipolar disorder, the exploration of concomitant treatments are yet to be determined. Future studies
the molecular determinants putatively involved in the therapeutic effect combining tDCS with other interventions should not only focus on
of tDCS in bipolar depression is moving fast forward, building on pharmacotherapy but also on non-pharmacological trreatments, such as,
converging results from different areas of tDCS application. for example, cognitive-behavioral therapy (CBT), given the existing
evidence of psychotherapy efficacy in bipolar depression (Miklowitz
4.5. Study limitations et al., 2007) and of a synergistic action of tDCS and CBT in unipolar
depression (D’Urso et al., 2013). As regards the field of neurobiological
Among the limitations of this review, data on patients demographics, studies, it would be interesting to explore commonalities and differences
onset and course of illness were lacking in detail. Moreover, we have not between the mechanisms involved in pharmacological interventions and
conducted any comparison between the daily and the twice-a-day ses­ non-invasive brain stimulation treatments as well as among different
sions frequency, because of the great disparity in the respective number non-invasive brain stimulation techniques.
of studies (in favor of daily sessions) and of too many concomitant
variables potentially influencing the tDCS response. Finally, the sample 5. Conclusions
size of bipolar depressed patients was small. However, this is in line with
the paucity of literature on tDCS across bipolar disorders. Even more The present systematic review examined the efficacy, safety, and
sources of variability are present in the analyzed neurobiological liter­ putative neurobiological underpinnings of tDCS in bipolar depression,
ature. Due to this remarkable lack of uniformity among studies and the suggesting that the technique might be a promising therapeutic mo­
overall paucity of data regarding tDCS specifically for bipolar depres­ dality for this condition, even more than for unipolar depression.
sion, we chose to perform a systematic review and not to adopt a meta- Further randomized controlled trials and more specific neurobiological
analytic approach. studies are required to definitively conclude whether tDCS has utility for
the acute treatment or the prevention of bipolar depression.
4.6. Implications for future research
Role of the funding source
In the last two decades, the research on rTMS as a treatment for
depression has been growing more and more, leading to the clearence of This research did not receive any specific grant from funding
this therapy by several national regulatory entities. On the contrary, the agencies in the public, commercial, or not-for-profit sectors.
less investigated tDCS has not yet been accepted as an official treatment
of depression, mainly because of insufficient evidence of efficacy. In our
opinion, further research on tDCS for depression, and particularly for Declaration of Competing Interest
bipolar depression, is justified by some important advantages of this
technique over rTMS. Firstly, tDCS is safer, showing much less risk of GDU, ET, AB, MP, BDO, GDL, AM, GM, MF, FI have no interest to
seizures, and has smoother side effects, i.e., mostly itching and tingling declare; AdB has received research support from Janssen, Lundbeck, and
at the site of stimulation, while TMS can be painful and induce strong Otsuka and lecture fees for educational meeting from Chiesi, Lundbeck,
discomfort to the patient’s head. Secondly, tDCS is more feasible, given Roche, Sunovion, Vitria, Recordati, Angelini and Takeda; he has served
that is cheaper and easier to administer, i.e., even by non-health pro­ on advisory boards for Eli Lilly, Jansen, Lundbeck, Otsuka, Roche, and
fessional, if adequately trained. Moreover, the portability of the tDCS Takeda, Chiesi, Recordati, Angelini, Vitria.
device makes it possible a home-based treatment, which entails less
burden on daily routines of patients and their families. In addition, it Appendix A. Supplementary data
does not require immobility, which is a better option for restless patients
such as those with depression with mixed features and allows the Supplementary data to this article can be found online at https://doi.
concomitant administration of cognitive training and/or psychotherapy. org/10.1016/j.pnpbp.2022.110672.
Finally, in research settings, it is more suitable for double-blind and
sham-controlled studies, being the allocation concealment easier to References
perform.
Further randomized controlled trials specifically targeted to patients Aguilar-Valles, A., Rodrigue, B., Matta-Camacho, E., 2020. Maternal immune activation
with bipolar depression or, in alternative, clearly separating the effects and the development of dopaminergic neurotran a smission of the offspring:
relevance for schizophrenia and other psychoses. Front. Psychiatry 11, 852. https://
of tDCS on unipolar and bipolar depressed patients are needed. More­ doi.org/10.3389/fpsyt.2020.00852.
over, larger samples are warranted to confirm the described preliminary Akula, N., Wendland, J.R., Choi, K.H., McMahon, F.J., 2016. An Integrative Genomic
findings and to ascertain the most beneficial tDCS treatment protocols. Study Implicates the Postsynaptic Density in the Pathogenesis of Bipolar Disorder.
Neuropsychopharmacology 41, 886–895. https://doi.org/10.1038/npp.2015.218.
Future trials are also needed to eliminate the sham tDCS placebo anti­ Alfieri, A., Sorokina, O., Adrait, A., Angelini, C., Russo, I., Morellato, A., Matteoli, M.,
depressant effect, for which there is important evidence (Loo et al., Menna, E., Boeri Erba, E., McLean, C., Armstrong, J.D., Ala, U., Buxbaum, J.D.,
2012, 2018). In addition, one of the most interesting areas to explore in Brusco, A., Couté, Y., De Rubeis, S., Turco, E., Defilippi, P., 2017. Synaptic
interactome mining reveals p140Cap as a new hub for PSD proteins involved in
the future studies is the identification of response predictors. In fact, a
psychiatric and neurological disorders. Front. Mol. Neurosci. 10, 212. https://doi.
work from our group (D’Urso et al., 2017) we investigated which spe­ org/10.3389/fnmol.2017.00212.
cific clinical characteristics were predictive of the acute antidepressant Alvarez-Alvarado, S., Boutzoukas, E.M., Kraft, J.N., O'Shea, A., Indahlastari, A.,
effect of bifrontal tDCS in a sample of 171 depressed patients. Indeed, we Albizu, A., Nissim, N.R., Evangelista, N.D., Cohen, R., Porges, E.C., Woods, A.J.,
2021. Impact of transcranial direct current stimulation and cognitive training on
found that the reduction in HDRS scores after tDCS was strongly asso­ frontal lobe neurotransmitter concentrations. Front. Aging Neurosci. 13, 761348
ciated with the baseline values of “Cognitive Disturbances” and https://doi.org/10.3389/fnagi.2021.761348.

14
G. D'Urso et al. Progress in Neuropsychopharmacology & Biological Psychiatry 121 (2023) 110672

Aparicio, L.V.M., Rosa, V., Razza, L.M., Sampaio-Junior, B., Borrione, L., Valiengo, L., Brunoni, A.R., Machado-Vieira, R., Zarate, C.A., Valiengo, L., Vieira, E.L., Benseñor, I.M.,
Lotufo, P.A., Benseñor, I.M., Fraguas, R., Moffa, A.H., Gattaz, W.F., Brunoni, A.R., Lotufo, P.A., Gattaz, W.F., Teixeira, A.L., 2014. Cytokines plasma levels during
2019. Transcranial direct current stimulation (tDCS) for preventing major depressive antidepressant treatment with sertraline and transcranial direct current stimulation
disorder relapse: Results of a 6-month follow-up. Depress. Anxiety 36, 262–268. (tDCS): results from a factorial, randomized, controlled trial. Psychopharmacology
https://doi.org/10.1002/da.22878. 231, 1315–1323. https://doi.org/10.1007/s00213-013-3322-3.
Ashok, A.H., Marques, T.R., Jauhar, S., Nour, M.M., Goodwin, G.M., Young, A.H., Brunoni, A.R., Moffa, A.H., Fregni, F., Palm, U., Padberg, F., Blumberger, D.M.,
Howes, O.D., 2017. The dopamine hypothesis of bipolar affective disorder: the state Daskalakis, Z.J., Bennabi, D., Haffen, E., Alonzo, A., Loo, C.K., 2016. Transcranial
of the art and implications for treatment. Mol. Psychiatry 22, 666–679. https://doi. direct current stimulation for acute major depressive episodes: meta-analysis of
org/10.1038/mp.2017.16. individual patient data. Br. J. Psychiatry 208, 522–531. https://doi.org/10.1192/
Bachen, E.A., Chesney, M.A., Criswell, L.A., 2009. Prevalence of mood and anxiety bjp.bp.115.164715.
disorders in women with systemic lupus erythematosus. Arthritis Rheum. 61, Buccelli, C., Di Lorenzo, P., Paternoster, M., D'Urso, G., Graziano, V., Niola, M., 2016.
822–829. https://doi.org/10.1002/art.24519. Electroconvulsive Therapy in Italy: Will Public Controversies Ever Stop? J. ECT 32,
Bachtiar, V., Johnstone, A., Berrington, A., Lemke, C., Johansen-Berg, H., Emir, U., 207–211. https://doi.org/10.1097/yct.0000000000000301.
Stagg, C.J., 2018. Modulating regional motor cortical excitability with noninvasive Bunai, T., Hirosawa, T., Kikuchi, M., Fukai, M., Yokokura, M., Ito, S., Takata, Y.,
brain stimulation results in neurochemical changes in bilateral motor cortices. Terada, T., Ouchi, Y., 2021. tDCS-induced modulation of GABA concentration and
J. Neurosci. 38 (33), 7327–7336. https://doi.org/10.1523/JNEUROSCI.2853- dopamine release in the human brain: A combination study of magnetic resonance
17.2018. spectroscopy and positron emission tomography. Brain Stimulation 14, 154–160.
Bai, S., Dokos, S., Ho, K.-A., Loo, C., 2014. A computational modelling study of https://doi.org/10.1016/j.brs.2020.12.010.
transcranial direct current stimulation montages used in depression. NeuroImage 87, Callai, E.M.M., Zin, L.E.F., Catarina, L.S., Ponzoni, D., Gonçalves, C.A.S., Vizuete, A.F.K.,
332–344. https://doi.org/10.1016/j.neuroimage.2013.11.015. Cougo, M.C., Boff, J., Puricelli, E., Fernandes, E.K., da Silva Torres, I.L., Quevedo, A.
Baliga, S., Sreeraj, V.S., Parlikar, R., Rai, D., Chhabra, H., Kumar, V., S., 2022. Evaluation of the immediate effects of a single transcranial direct current
Venkatasubramanian, G., 2020. Role of transcranial direct current stimulation in stimulation session on astrocyte activation, inflammatory response, and pain
bipolar depression: A case report. Asian J. Psychiatr. 47, 101873. https://doi.org/ threshold in naïve rats. Behav. Brain Res. 428, 113880 https://doi.org/10.1016/j.
10.1016/j.ajp.2019.101873. bbr.2022.113880.
Barbati, S.A., Cocco, S., Longo, V., Spinelli, M., Gironi, K., Mattera, A., Paciello, F., Cancel, L.M., Arias, K., Bikson, M., Tarbell, J.M., 2018. Direct current stimulation of
Colussi, C., Podda, M.V., Grassi, C., 2020. Enhancing Plasticity Mechanisms in the endothelial monolayers induces a transient and reversible increase in transport due
Mouse Motor Cortex by Anodal Transcranial Direct-Current Stimulation: The to the electroosmotic effect. Sci. Rep. 8, 9265. https://doi.org/10.1038/s41598-018-
Contribution of Nitric Oxide Signaling. Cerebral cortex, 30, pp. 2972–2985. https:// 27524-9.
doi.org/10.1093/cercor/bhz288. Cattaneo, C.I., Ressico, F., Fornaro, M., Fazzari, G., Perugi, G., 2022. The shocking
Barone, A., Signoriello, S., Latte, G., Vellucci, L., Giordano, G., Avagliano, C., attitude toward electroconvulsive therapy in Italy. CNS Spectrums 27, 131–133.
Buonaguro, E.F., Marmo, F., Tomasetti, C., Iasevoli, F., de Bartolomeis, A., 2021. https://doi.org/10.1017/s1092852920002059.
Modulation of glutamatergic functional connectivity by a prototypical antipsychotic: Celano, E., Tiraboschi, E., Consogno, E., D'Urso, G., Mbakop, M.P., Gennarelli, M., de
Translational inference from a postsynaptic density immediate-early gene-based Bartolomeis, A., Racagni, G., Popoli, M., 2003. Selective regulation of presynaptic
network analysis. Behav. Brain Res. 404, 113160 https://doi.org/10.1016/j. calcium/calmodulin-dependent protein kinase II by psychotropic drugs. Biol.
bbr.2021.113160. Psychiatry 53, 442–449. https://doi.org/10.1016/s0006-3223(02)01491-9.
Beck, A.T., Steer, R.A., Brown, G.K.J.S.A., 1996. Beck depression inventory manual. Chen, M., Jiang, Q., Zhang, L., 2021. The prevalence of bipolar disorder in autoimmune
Psychol. Corp. 785–791. disease: a systematic review and meta-analysis. Ann. Palliative Med. 10, 350–361.
Bersani, F.S., Minichino, A., Bernabei, L., Spagnoli, F., Corrado, A., Vergnani, L., https://doi.org/10.21037/apm-20-2293.
Mannarelli, D., Pauletti, C., Fattapposta, F., Biondi, M., et al., 2016. Prefronto- Chew, T., Ho, K.-A., Loo, C.K., 2015. Inter- and Intra-individual Variability in Response
cerebellar tDCS enhances neurocognition in euthymic bipolar patients. Findings to Transcranial Direct Current Stimulation (tDCS) at Varying Current Intensities.
from a placebo-controlled neuropsychological and psychophysiological Brain Stimulation 8, 1130–1137. https://doi.org/10.1016/j.brs.2015.07.031.
investigation. J. Affect. Disord. 209, 262–269. https://doi.org/10.1016/j. Chhabra, H., Shivakumar, V., Subbanna, M., Kalmady, S.V., Bose, A., Agarwal, S.M.,
jad.2016.11.037. Sreeraj, V.S., Dinakaran, D., Narayanaswamy, J.C., Debnath, M.,
Bersani, F.S., Minichino, A., Fattapposta, F., Bernabei, L., Spagnoli, F., Mannarelli, D., Venkatasubramanian, G., 2018. Gene polymorphisms and response to transcranial
Francesconi, M., Pauletti, C., Corrado, A., Vergnani, L., et al., 2015. direct current stimulation for auditory verbal hallucinations in schizophrenia. Acta
Prefrontocerebellar transcranial direct current stimulation increases amplitude and Neuropsychiatrica 30, 218–225. https://doi.org/10.1017/neu.2018.4.
decreases latency of P3b component in patients with euthymic bipolar disorder. Chiou, Y.-J., Huang, T.-L., 2019. Brain-derived neurotrophic factor (BDNF) and bipolar
Neuropsychiatr. Dis. Treat. 11, 2913–2917. https://doi.org/10.2147/ndt.s91625. disorder. Psychiatry Res. 274, 395–399. https://doi.org/10.1016/j.
Bikson, M., Bulow, P., Stiller, J.W., Datta, A., Battaglia, F., Karnup, S.V., Postolache, T.T., psychres.2019.02.051.
2008. Transcranial direct current stimulation for major depression: A general system Chou, Y.H., Wang, S.J., Lin, C.L., Mao, W.C., Lee, S.M., Liao, M.H., 2010. Decreased brain
for quantifying transcranial electrotherapy dosage. Curr. Treat. Options Neurol. 10, serotonin transporter binding in the euthymic state of bipolar I but not bipolar II
377. https://doi.org/10.1007/s11940-008-0040-y. disorder: a SPECT study. Bipolar Disord. 12, 312–318. https://doi.org/10.1111/
Bikson, M., Grossman, P., Thomas, C., Zannou, A.L., Jiang, J., Adnan, T., j.1399-5618.2010.00800.x.
Mourdoukoutas, A.P., Kronberg, G., Truong, D., Boggio, P., Brunoni, A.R., Cirillo, G., Di Pino, G., Capone, F., Ranieri, F., Florio, L., Todisco, V., Tedeschi, G.,
Charvet, L., Fregni, F., Fritsch, B., Gillick, B., Hamilton, R.H., Hampstead, B.M., Funke, K., Di Lazzaro, V., 2017. Neurobiological after-effects of non-invasive brain
Jankord, R., Kirton, A., Knotkova, H., Liebetanz, D., Liu, A., Loo, C., Nitsche, M.A., stimulation. Brain Stimulation 10, 1–18. https://doi.org/10.1016/j.
Reis, J., Richardson, J.D., Rotenberg, A., Turkeltaub, P.E., Woods, A.J., 2016. Safety brs.2016.11.009.
of Transcranial Direct Current Stimulation: Evidence Based Update 2016. Brain Corsi-Zuelli, F., Deakin, B., 2021. Impaired regulatory T cell control of astroglial
Stimulation 9, 641–661. https://doi.org/10.1016/j.brs.2016.06.004. overdrive and microglial pruning in schizophrenia. Neurosci. Biobehav. Rev. 125,
Boggio, P.S., Rigonatti, S.P., Ribeiro, R.B., Myczkowski, M.L., Nitsche, M.A., Pascual- 637–653. https://doi.org/10.1016/j.neubiorev.2021.03.004.
Leone, A., Fregni, F., 2008. A randomized, double-blind clinical trial on the efficacy D’Urso, G., Mantovani, A., Micillo, M., Priori, A., Muscettola, G., 2013. Transcranial
of cortical direct current stimulation for the treatment of major depression. Int. J. direct current stimulation and cognitive-behavioral therapy: evidence of a
Neuropsychopharmacol. 11, 249–254. https://doi.org/10.1017/ synergistic effect in treatment-resistant depression. Brain Stimulation 6, 465–467.
S1461145707007833. https://doi.org/10.1016/j.brs.2012.09.003.
Brooks, J.O., Hoblyn, J.C., Ketter, T.A., 2010. Metabolic evidence of corticolimbic D’Urso, G., Dell’Osso, B., Rossi, R., Brunoni, A.R., Bortolomasi, M., Ferrucci, R.,
dysregulation in bipolar mania. Psychiatry Res. Neuroimaging 181, 136–140. Priori, A., de Bartolomeis, A., Altamura, A.C., 2017. Clinical predictors of acute
https://doi.org/10.1016/j.pscychresns.2009.08.006. response to transcranial direct current stimulation (tDCS) in major depression.
Brunoni, A.R., Ferrucci, R., Bortolomasi, M., Vergari, M., Tadini, L., Boggio, P.S., J. Affect. Disord. 219, 25–30. https://doi.org/10.1016/j.jad.2017.05.019.
Giacopuzzi, M., Barbieri, S., Priori, A., 2011a. Transcranial direct current stimulation D'Addario, C., Palazzo, M.C., Benatti, B., Grancini, B., Pucci, M., Di Francesco, A.,
(tDCS) in unipolar vs. bipolar depressive disorder. Prog. Neuro-Psychopharmacol. Camuri, G., Galimberti, D., Fenoglio, C., Scarpini, E., Altamura, A.C.,
Biol. Psychiatry 35, 96–101. https://doi.org/10.1016/j.pnpbp.2010.09.010. Maccarrone, M., Dell'Osso, B., 2018. Regulation of gene transcription in bipolar
Brunoni, A.R., Valiengo, L., Zanao, T., de Oliveira, J.F., Bensenor, I.M., Fregni, F., 2011b. disorders: Role of DNA methylation in the relationship between prodynorphin and
Manic psychosis after sertraline and transcranial direct-current stimulation. brain derived neurotrophic factor. Prog. Neuro-Psychopharmacol. Biol. Psychiatry
J. Neuropsychiatry Clin Neurosci. Summer 23 (3), E4–E5. https://doi.org/10.1176/ 82, 314–321. https://doi.org/10.1016/j.pnpbp.2017.08.011.
jnp.23.3.jnpe4. Datta, A., Elwassif, M., Battaglia, F., Bikson, M., 2008. Transcranial current stimulation
Brunoni, A.R., Ferrucci, R., Bortolomasi, M., Scelzo, E., Boggio, P.S., Fregni, F., focality using disc and ring electrode configurations: FEM analysis. J. Neural Eng. 5,
Dell'Osso, B., Giacopuzzi, M., Altamura, A.C., Priori, A., 2013a. Interactions between 163–174. https://doi.org/10.1088/1741-2560/5/2/007.
transcranial direct current stimulation (tDCS) and pharmacological interventions in de Bartolomeis, A., Tomasetti, C., 2012. Calcium-dependent networks in
the Major Depressive Episode: findings from a naturalistic study. Eur. Psychiatry 28, dopamine–glutamate interaction: the role of postsynaptic scaffolding proteins. Mol.
356–361. https://doi.org/10.1016/j.eurpsy.2012.09.001. Neurobiol. 46, 275–296. https://doi.org/10.1007/s12035-012-8293-6.
Brunoni, A.R., Kemp, A.H., Shiozawa, P., Cordeiro, Q., Valiengo, L.C.L., Goulart, A.C., de Bartolomeis, A., Tomasetti, C., Cicale, M., Yuan, P.X., Manji, H.K., 2012. Chronic
Coprerski, B., Lotufo, P.A., Brunoni, D., Perez, A.B.A., Fregni, F., Benseñor, I.M., treatment with lithium or valproate modulates the expression of Homer1b/c and its
2013b. Impact of 5-HTTLPR and BDNF polymorphisms on response to sertraline related genes Shank and Inositol 1,4,5-trisphosphate receptor. Eur.
versus transcranial direct current stimulation: Implications for the serotonergic Neuropsychopharmacol. 22, 527–535. https://doi.org/10.1016/j.
system. Eur. Neuropsychopharmacol. 23, 1530–1540. https://doi.org/10.1016/j. euroneuro.2011.11.006.
euroneuro.2013.03.009.

15
G. D'Urso et al. Progress in Neuropsychopharmacology & Biological Psychiatry 121 (2023) 110672

de Bartolomeis, A., Avvisati, L., Iasevoli, F., Tomasetti, C., 2013. Intracellular pathways implicates synaptic function and energy pathways in bipolar disorder. Transl.
of antipsychotic combined therapies: implication for psychiatric disorders treatment. Psychiatry 6, e959. https://doi.org/10.1038/tp.2016.224.
Eur. J. Pharmacol. 718, 502–523. https://doi.org/10.1016/j.ejphar.2013.06.034. Fonteneau, C., Redoute, J., Haesebaert, F., Le Bars, D., Costes, N., Suaud-Chagny, M.F.,
de Bartolomeis, A., Buonaguro, E.F., Iasevoli, F., Tomasetti, C., 2014a. The emerging role Brunelin, J., 2018. Frontal transcranial direct current stimulation induces dopamine
of dopamine-glutamate interaction and of the postsynaptic density in bipolar release in the ventral striatum in human. Cereb. Cortex 28, 2636–2646. https://doi.
disorder pathophysiology: Implications for treatment. J. Psychopharmacol. 28, org/10.1093/cercor/bhy093.
505–526. https://doi.org/10.1177/0269881114523864. Fonteneau, C., Mondino, M., Arns, M., Baeken, C., Bikson, M., Brunoni, A.R., Burke, M.J.,
de Bartolomeis, A., Latte, G., Tomasetti, C., Iasevoli, F., 2014b. Glutamatergic Neuvonen, T., Padberg, F., Pascual-Leone, A., Poulet, E., Ruffini, G.,
postsynaptic density protein dysfunctions in synaptic plasticity and dendritic spines Santarnecchi, E., Sauvaget, A., Schellhorn, K., Suaud-Chagny, M.F., Palm, U.,
morphology: relevance to schizophrenia and other behavioral disorders Brunelin, J., 2019. Sham tDCS: A hidden source of variability? Reflections for further
pathophysiology, and implications for novel therapeutic approaches. Mol. blinded, controlled trials. Brain Stimul. 12 (3), 668–673. https://doi.org/10.1016/j.
Neurobiol. 49, 484–511. https://doi.org/10.1007/s12035-013-8534-3. brs.2018.12.977 (Epub 2019 Jan 2. PMID: 30639235).
de Bartolomeis, A., Barone, A., Begni, V., Riva, M.A., 2022a. Present and future Fornaro, M., Stubbs, B., De Berardis, D., Perna, G., Valchera, A., Veronese, N., Solmi, M.,
antipsychotic drugs: A systematic review of the putative mechanisms of action for Ganança, L., 2016. Atypical antipsychotics in the treatment of acute bipolar
efficacy and a critical appraisal under a translational perspective. Pharmacol. Res. depression with mixed features: a systematic review and exploratory meta-analysis
176, 106078 https://doi.org/10.1016/j.phrs.2022.106078. of placebo-controlled clinical trials. Int. J. Mol. Sci. 17, 241. https://doi.org/
de Bartolomeis, A., Barone, A., Buonaguro, E.F., Tomasetti, C., Vellucci, L., Iasevoli, F., 10.3390/ijms17020241.
2022b. The Homer1 family of proteins at the crossroad of dopamine-glutamate Fornaro, M., De Berardis, D., Perna, G., Solmi, M., Veronese, N., Orsolini, L.,
signaling: An emerging molecular "Lego" in the pathophysiology of psychiatric Buonaguro, E.F., Iasevoli, F., Köhler, C.A., Carvalho, A.F., de Bartolomeis, A., 2017.
disorders. A systematic review and translational insight. Neurosci. Biobehav. Rev. Lurasidone in the treatment of bipolar depression: systematic review of systematic
136, 104596 https://doi.org/10.1016/j.neubiorev.2022.104596. reviews. Biomed. Res. Int. 2017, 3084859. https://doi.org/10.1155/2017/3084859.
De Berardis, D., Orsolini, L., Iasevoli, F., Prinzivalli, E., de Bartolomeis, A., Serroni, N., Fornaro, M., Anastasia, A., Novello, S., Fusco, A., Solmi, M., Monaco, F., Veronese, N., De
Mazza, M., Valchera, A., Fornaro, M., Vecchiotti, R., Carano, A., Sepede, G., Berardis, D., de Bartolomeis, A., 2018. Incidence, prevalence and clinical correlates
Vellante, F., Matarazzo, I., Pompili, M., Perna, G., Conti, C., Segura-García, C., of antidepressant-emergent mania in bipolar depression: a systematic review and
Martinotti, G., Di Giannantonio, M., 2016. The Novel Antipsychotic Cariprazine meta-analysis. Bipolar Disord. 20, 195–227. https://doi.org/10.1111/bdi.12612.
(RGH-188): State-of-the-Art in the Treatment of Psychiatric Disorders. Curr. Pharm. Fornaro, M., Carvalho, A.F., Fusco, A., Anastasia, A., Solmi, M., Berk, M., Sim, K.,
Des. 22, 5144–5162. https://doi.org/10.2174/1381612822666160701084447. Vieta, E., de Bartolomeis, A., 2020. The concept and management of acute episodes
De-Paula, V.J., Gattaz, W.F., Forlenza, O.V., 2016. Long-term lithium treatment increases of treatment-resistant bipolar disorder: a systematic review and exploratory meta-
intracellular and extracellular brain-derived neurotrophic factor (BDNF) in cortical analysis of randomized controlled trials. J. Affect. Disord. 276, 970–983. https://doi.
and hippocampal neurons at subtherapeutic concentrations. Bipolar Disord. 18, org/10.1016/j.jad.2020.07.109.
692–695. https://doi.org/10.1111/bdi.12449. Fresnoza, S.M., Batsikadze, G., Müller, L.E., Rost, C., Chamoun, M., Paulus, W., Kuo, M.
Dondé, C., Amad, A., Nieto, I., Brunoni, A.R., Neufeld, N.H., Bellivier, F., Poulet, E., F., Nitsche, M.A., 2021. Inhibitory effect of apomorphine on focal and nonfocal
Geoffroy, P.A., 2017. Transcranial direct-current stimulation (tDCS) for bipolar plasticity in the human motor cortex. Pharmaceutics 13. https://doi.org/10.3390/
depression: A systematic review and meta-analysis. Prog. Neuro-Psychopharmacol. pharmaceutics13050718.
Biol. Psychiatry 78, 123–131. https://doi.org/10.1016/j.pnpbp.2017.05.021. Fukai, M., Bunai, T., Hirosawa, T., Kikuchi, M., Ito, S., Minabe, Y., Ouchi, Y., 2019.
Dondé, C., Neufeld, N.H., Geoffroy, P.A., 2018. The impact of transcranial direct current Endogenous dopamine release under transcranial direct-current stimulation governs
stimulation (tDCS) on bipolar depression, mania, and euthymia: a systematic review enhanced attention: a study with positron emission tomography. Transl. Psychiatry
of preliminary data. Psychiatr Q. 89 (4), 855–867. https://doi.org/10.1007/s11126- 9, 115. https://doi.org/10.1038/s41398-019-0443-4.
018-9584-5. Gallo, F.T., Katche, C., Morici, J.F., Medina, J.H., Weisstaub, N.V., 2018. Immediate
D’Urso, G., Ferrucci, R., Bruzzese, D., Pascotto, A., Priori, A., Altamura, C.A., Early Genes, Memory and Psychiatric Disorders: Focus on c-Fos. Egr1 and Arc 12.
Galderisi, S., Bravaccio, C., 2014. Transcranial direct current stimulation for autistic https://doi.org/10.3389/fnbeh.2018.00079.
disorder. Biol. Psychiatry 76, e5–e6. https://doi.org/10.1016/j. Gálvez, V., Alonzo, A., Martin, D., Mitchell, P.B., Sachdev, P., Loo, C.K., 2011.
biopsych.2013.11.009. Hypomania induction in a patient with bipolar II disorder by transcranial direct
D'Urso, G., Bruzzese, D., Ferrucci, R., Priori, A., Pascotto, A., Galderisi, S., Altamura, A. current stimulation (tDCS). J ECT. 27 (3), 256–258. https://doi.org/10.1097/
C., Bravaccio, C., 2015. Transcranial direct current stimulation for hyperactivity and YCT.0b013e3182012b89.
noncompliance in autistic disorder. World J. Biol. Psychiatry 16, 361–366. https:// Gao, J., Jia, M., Qiao, D., Qiu, H., Sokolove, J., Zhang, J., Pan, Z., 2016. TPH2 gene
doi.org/10.3109/15622975.2015.1014411. polymorphisms and bipolar disorder: A meta-analysis. Am. J. Med. Genet. B
D'Urso, G., Brunoni, A.R., Mazzaferro, M.P., Anastasia, A., de Bartolomeis, A., Neuropsychiatr. Genet. 171b, 145–152. https://doi.org/10.1002/ajmg.b.32381.
Mantovani, A., 2016. Transcranial direct current stimulation for obsessive- Geddes, J.R., Calabrese, J.R., Goodwin, G.M., 2009. Lamotrigine for treatment of bipolar
compulsive disorder: A randomized, controlled, partial crossover trial. Depress. depression: independent meta-analysis and meta-regression of individual patient
Anxiety 33, 1132–1140. https://doi.org/10.1002/da.22578. data from five randomised trials. Br. J. Psychiatry 194, 4–9. https://doi.org/
D'Urso, G., Mantovani, A., Patti, S., Toscano, E., de Bartolomeis, A., 2018. Transcranial 10.1192/bjp.bp.107.048504.
direct current stimulation in obsessive-compulsive disorder, posttraumatic stress Ghanavati, E., Salehinejad, M.A., De Melo, L., Nitsche, M.A., Kuo, M.F., 2022. NMDA
disorder, and anxiety disorders. J. ECT 34, 172–181. https://doi.org/10.1097/ receptor-related mechanisms of dopaminergic modulation of tDCS-induced
yct.0000000000000538. neuroplasticity. Cereb. Cortex. https://doi.org/10.1093/cercor/bhac028.
D’Urso, G., Toscano, E., Sanges, V., Sauvaget, A., Sheffer, C.E., Riccio, M.P., Ferrucci, R., Goerigk, S., Cretaz, E., Sampaio-Junior, B., Vieira, É., Gattaz, W., Klein, I., Lafer, B.,
Iasevoli, F., Priori, A., Bravaccio, C., de Bartolomeis, A., 2021. Cerebellar Teixeira, A.L., Carvalho, A.F., Lotufo, P.A., Benseñor, I.M., Bühner, M., Padberg, F.,
transcranial direct current stimulation in children with autism spectrum disorder: a Brunoni, A.R., 2021. Effects of tDCS on neuroplasticity and inflammatory biomarkers
pilot study on efficacy, feasibility, safety, and unexpected outcomes in tic disorder in bipolar depression: Results from a sham-controlled study. Prog. Neuro-
and epilepsy. J. Clin. Med. 11 (1), 143. https://doi.org/10.3390/jcm11010143. Psychopharmacol. Biol. Psychiatry 105, 110119. https://doi.org/10.1016/j.
Farhi, A., Cohen, A.D., Shovman, O., Comaneshter, D., Amital, H., Amital, D., 2016. pnpbp.2020.110119.
Bipolar disorder associated with rheumatoid arthritis: A case-control study. J. Affect. Grewal, S., McKinlay, S., Kapczinski, F., Pfaffenseller, B., Wollenhaupt-Aguiar, B., 2022.
Disord. 189, 287–289. https://doi.org/10.1016/j.jad.2015.09.058. Biomarkers of neuroprogression and late staging in bipolar disorder: A systematic
Ferrari, A.J., Stockings, E., Khoo, J.P., Erskine, H.E., Degenhardt, L., Vos, T., review. Austr. New Zeal. J. Psychiatry. https://doi.org/10.1177/
Whiteford, H.A., 2016. The prevalence and burden of bipolar disorder: findings from 00048674221091731, 48674221091731.
the Global Burden of Disease Study 2013. Bipolar Disord. 18, 440–450. https://doi. Grimm, S., Beck, J., Schuepbach, D., Hell, D., Boesiger, P., Bermpohl, F., Niehaus, L.,
org/10.1111/bdi.12423. Boeker, H., Northoff, G., 2008. Imbalance between left and right dorsolateral
Ferrucci, R., Bortolomasi, M., Brunoni, A., Vergari, M., Tadini, L., Giacopuzzi, M., prefrontal cortex in major depression is linked to negative emotional judgment: an
Priori, A., 2009. Comparative benefits of transcranial direct current stimulation fMRI study in severe major depressive disorder. Biol. Psychiatry 63, 369–376.
(TDCS) treatment in patients with mild/moderate vs. severe depression. https://doi.org/10.1016/j.biopsych.2007.05.033.
Ferrucci, R., Mrakic-Sposta, S., Gardini, S., Ruggiero, F., Vergari, M., Mameli, F., Gross, M., Nakamura, L., Pascual-Leone, A., Fregni, F., 2007. Has repetitive transcranial
Arighi, A., Spallazzi, M., Barocco, F., Michelini, G., Pietroboni, A.M., Ghezzi, L., magnetic stimulation (rTMS) treatment for depression improved? A systematic
Fumagalli, G.G., D'Urso, G., Caffarra, P., Scarpini, E., Priori, A., Marceglia, S., 2018. review and meta-analysis comparing the recent vs. the earlier rTMS studies. Acta
Behavioral and neurophysiological effects of transcranial direct current stimulation Psychiatr. Scand. 116, 165–173. https://doi.org/10.1111/j.1600-0447.2007.01049.
(tDCS) in fronto-temporal dementia. Front. Behav. Neurosci. 12, 235. https://doi. x.
org/10.3389/fnbeh.2018.00235. Grunze, H., Vieta, E., Goodwin, G.M., Bowden, C., Licht, R.W., Möller, H.J., Kasper, S.,
Filmer, H.L., Ehrhardt, S.E., Bollmann, S., Mattingley, J.B., Dux, P.E., 2019. Accounting 2013. The World Federation of Societies of Biological Psychiatry (WFSBP) guidelines
for individual differences in the response to tDCS with baseline levels of for the biological treatment of bipolar disorders: update 2012 on the long-term
neurochemical excitability. Cortex 115, 324–334. https://doi.org/10.1016/j. treatment of bipolar disorder. World J. Biol. Psychiatry 14, 154–219. https://doi.
cortex.2019.02.012. org/10.3109/15622975.2013.770551.
Flöel, A., 2014. tDCS-enhanced motor and cognitive function in neurological diseases. Halff, E.F., Cotel, M.C., Natesan, S., McQuade, R., Ottley, C.J., Srivastava, D.P.,
NeuroImage 85 (Pt 3), 934–947. https://doi.org/10.1016/j. Howes, O.D., Vernon, A.C., 2021. Effects of chronic exposure to haloperidol,
neuroimage.2013.05.098. olanzapine or lithium on SV2A and NLGN synaptic puncta in the rat frontal cortex.
Föcking, M., Dicker, P., Lopez, L.M., Hryniewiecka, M., Wynne, K., English, J.A., Behav. Brain Res. 405, 113203 https://doi.org/10.1016/j.bbr.2021.113203.
Cagney, G., Cotter, D.R., 2016. Proteomic analysis of the postsynaptic density Hamilton, M., 1960. A rating scale for depression. J. Neurol. Neurosurg. Psychiatry 23,
56–62. https://doi.org/10.1136/jnnp.23.1.56.

16
G. D'Urso et al. Progress in Neuropsychopharmacology & Biological Psychiatry 121 (2023) 110672

Higgins, J.P.T., Lasserson, T., Chandler, J., Tovey, D., Churchill, R., 2016. Transcranial direct current stimulation enhances neuroplasticity and accelerates
Methodological Expectations of Cochrane Intervention Reviews. Cochrane, London. motor recovery in a stroke mouse model. Stroke 53, 1746–1758. https://doi.org/
Hillen, A.E.J., Burbach, J.P.H., Hol, E.M., 2018. Cell adhesion and matricellular support 10.1161/strokeaha.121.034200.
by astrocytes of the tripartite synapse. Prog. Neurobiol. 165-167, 66–86. https://doi. Loo, C.K., Alonzo, A., Martin, D., Mitchell, P.B., Galvez, V., Sachdev, P., 2012.
org/10.1016/j.pneurobio.2018.02.002. Transcranial direct current stimulation for depression: 3-week, randomised, sham-
Ho, K.-A., Bai, S., Martin, D., Alonzo, A., Dokos, S., Loo, C.K., 2015. Clinical pilot study controlled trial. Br. J. Psychiatry 200, 52–59. https://doi.org/10.1192/bjp.
and computational modeling of bitemporal transcranial direct current stimulation, bp.111.097634.
and safety of repeated courses of treatment, in major depression. J ECT 31 (4), Loo, C.K., Husain, M.M., McDonald, W.M., Aaronson, S., O'Reardon, J.P., Alonzo, A.,
226–233. https://doi.org/10.1097/YCT.0000000000000230. Weickert, C.S., Martin, D.M., McClintock, S.M., Mohan, A., Lisanby, S.H., 2018.
Hsu, C.C., Chen, S.C., Liu, C.J., Lu, T., Shen, C.C., Hu, Y.W., Yeh, C.M., Chen, P.M., International randomized-controlled trial of transcranial Direct Current Stimulation
Chen, T.J., Hu, L.Y., 2014. Rheumatoid arthritis and the risk of bipolar disorder: a in depression. Brain Stimulation 11, 125–133. https://doi.org/10.1016/j.
nationwide population-based study. PLoS One 9, e107512. https://doi.org/10.1371/ brs.2017.10.011.
journal.pone.0107512. Lugon, M.D.M.V., Batsikadze, G., Fresnoza, S., Grundey, J., Kuo, M.-F., Paulus, W.,
Hunter, M.A., Coffman, B.A., Gasparovic, C., Calhoun, V.D., Trumbo, M.C., Clark, V.P., Nakamura-Palacios, E.M., Nitsche, M.A., 2017. Mechanisms of nicotinic modulation
2015. Baseline effects of transcranial direct current stimulation on glutamatergic of glutamatergic neuroplasticity in humans. Cereb. Cortex 27, 544–553. https://doi.
neurotransmission and large-scale network connectivity. Brain Res. 1594, 92–107. org/10.1093/cercor/bhv252.
https://doi.org/10.1016/j.brainres.2014.09.066. MacDonald, K., Krishnan, A., Cervenka, E., Hu, G., Guadagno, E., Trakadis, Y., 2019.
Iasevoli, F., Tomasetti, C., de Bartolomeis, A., 2013. Scaffolding proteins of the post- Biomarkers for major depressive and bipolar disorders using metabolomics: A
synaptic density contribute to synaptic plasticity by regulating receptor localization systematic review. Am. J. Med. Genet. B Neuropsychiatric Genet. 180, 122–137.
and distribution: relevance for neuropsychiatric diseases. Neurochem. Res. 38, 1–22. https://doi.org/10.1002/ajmg.b.32680.
https://doi.org/10.1007/s11064-012-0886-y. Mardani, P., Zolghadriha, A., Dadashi, M., Javdani, H., Mousavi, S.E., 2021. Effect of
Iasevoli, F., Buonaguro, E.F., Avagliano, C., Barone, A., Eramo, A., Vellucci, L., de medication therapy combined with transcranial direct current stimulation on
Bartolomeis, A., 2020. The effects of antipsychotics on the synaptic plasticity gene depression and response inhibition of patients with bipolar disorder type I: a clinical
homer1a depend on a combination of their receptor profile, dose, duration of trial. BMC Psychiatry 21, 579. https://doi.org/10.1186/s12888-021-03592-6.
treatment, and brain regions targeted. Int. J. Mol. Sci. 21 https://doi.org/10.3390/ Martin, D.M., Alonzo, A., Mitchell, P.B., Sachdev, P., Gálvez, V., Loo, C.K., 2011. Fronto-
ijms21155555. extracephalic transcranial direct current stimulation as a treatment for major
Imburgio, M.J., Ballard, H.K., Cornwall, A.C., Worthy, D.A., Bernard, J.A., Orr, J.M., depression: an open-label pilot study. J. Affect. Disord. 134, 459–463. https://doi.
2021. Preliminary effects of prefrontal tDCS on dopamine-mediated behavior and org/10.1016/j.jad.2011.05.018.
psychophysiology. Behav. Brain Res. 402, 113091 https://doi.org/10.1016/j. Martin, D.M., Alonzo, A., Ho, K.A., Player, M., Mitchell, P.B., Sachdev, P., Loo, C.K.,
bbr.2020.113091. 2013. Continuation transcranial direct current stimulation for the prevention of
Jongkees, B.J., Loseva, A.A., Yavari, F.B., Nitsche, M.A., Colzato, L.S., 2019. The COMT relapse in major depression. J. Affect. Disord. 144, 274–278. https://doi.org/
Val(158) Met polymorphism does not modulate the after-effect of tDCS on working 10.1016/j.jad.2012.10.012.
memory. Eur. J. Neurosci. 49, 263–274. https://doi.org/10.1111/ejn.14261. Martin, D.M., McClintock, S.M., Aaronson, S.T., Alonzo, A., Husain, M.M., Lisanby, S.H.,
Kennedy, S.H., Javanmard, M., Vaccarino, F.J., 1997. A review of functional McDonald, W.M., Mohan, A., Nikolin, S., O'Reardon, J., Weickert, C.S., Loo, C.K.,
neuroimaging in mood disorders: positron emission tomography and depression. 2018a. Pre-treatment attentional processing speed and antidepressant response to
Can. J. Psychiatr. 42, 467–475. https://doi.org/10.1177/070674379704200502. transcranial direct current stimulation: Results from an international randomized
Kerner, B., 2015. Toward a deeper understanding of the genetics of bipolar disorder. controlled trial. Brain Stimulation 11, 1282–1290. https://doi.org/10.1016/j.
Front. Psychiatry 6, 105. https://doi.org/10.3389/fpsyt.2015.00105. brs.2018.08.011.
Kim, M.S., Koo, H., Han, S.W., Paulus, W., Nitsche, M.A., Kim, Y.H., Yoon, J.A., Shin, Y.I., Martin, P.M., Stanley, R.E., Ross, A.P., Freitas, A.E., Moyer, C.E., Brumback, A.C.,
2017. Repeated anodal transcranial direct current stimulation induces neural Iafrati, J., Stapornwongkul, K.S., Dominguez, S., Kivimäe, S., Mulligan, K.A.,
plasticity-associated gene expression in the rat cortex and hippocampus. Restor. Pirooznia, M., McCombie, W.R., Potash, J.B., Zandi, P.P., Purcell, S.M., Sanders, S.J.,
Neurol. Neurosci. 35, 137–146. https://doi.org/10.3233/rnn-160689. Zuo, Y., Sohal, V.S., Cheyette, B.N.R., 2018b. DIXDC1 contributes to psychiatric
Kim, J., Farchione, T., Potter, A., Chen, Q., Temple, R., 2019. Esketamine for treatment- susceptibility by regulating dendritic spine and glutamatergic synapse density via
resistant depression - first FDA-approved antidepressant in a new class. N. Engl. J. GSK3 and Wnt/β-catenin signaling. Mol. Psychiatry 23, 467–475. https://doi.org/
Med. 381, 1–4. https://doi.org/10.1056/NEJMp1903305. 10.1038/mp.2016.184.
Konopaske, G.T., Lange, N., Coyle, J.T., Benes, F.M., 2014. Prefrontal cortical dendritic Martinotti, G., Lupi, M., Montemitro, C., Miuli, A., Di Natale, C., Spano, M.C.,
spine pathology in schizophrenia and bipolar disorder. JAMA Psychiatry 71, Mancini, V., Lorusso, M., Stigliano, G., Tambelli, A., Di Carlo, F., Di Caprio, L.,
1323–1331. https://doi.org/10.1001/jamapsychiatry.2014.1582. Fraticelli, S., Chillemi, E., Pettorruso, M., Sepede, G., di Giannantonio, M., 2019.
Kuo, M.F., Paulus, W., Nitsche, M.A., 2008. Boosting focally-induced brain plasticity by Transcranial direct current stimulation reduces craving in substance use disorders: a
dopamine. Cereb. Cortex 18, 648–651. https://doi.org/10.1093/cercor/bhm098. double-blind, placebo-controlled study. J. ECT 35, 207–211. https://doi.org/
Kuo, M.F., Paulus, W., Nitsche, M.A., 2014. Therapeutic effects of non-invasive brain 10.1097/yct.0000000000000580.
stimulation with direct currents (tDCS) in neuropsychiatric diseases. NeuroImage 85 McClintock, S.M., Martin, D.M., Lisanby, S.H., Alonzo, A., McDonald, W.M., Aaronson, S.
(Pt 3), 948–960. https://doi.org/10.1016/j.neuroimage.2013.05.117. T., Husain, M.M., O'Reardon, J.P., Weickert, C.S., Mohan, A., Loo, C.K., 2020.
Kuo, H.-I., Paulus, W., Batsikadze, G., Jamil, A., Kuo, M.-F., Nitsche, M.A., 2016. Chronic Neurocognitive effects of transcranial direct current stimulation (tDCS) in unipolar
Enhancement of Serotonin Facilitates Excitatory Transcranial Direct Current and bipolar depression: Findings from an international randomized controlled trial.
Stimulation-Induced Neuroplasticity. Neuropsychopharmacology 41, 1223–1230. Depress. Anxiety 37, 261–272. https://doi.org/10.1002/da.22988.
https://doi.org/10.1038/npp.2015.270. McCutcheon, R.A., Brown, K., Nour, M.M., Smith, S.M., Veronese, M., Zelaya, F.,
Kupka, R.W., Nolen, W.A., Post, R.M., McElroy, S.L., Altshuler, L.L., Denicoff, K.D., Osugo, M., Jauhar, S., Hallett, W., Mehta, M.M., Howes, O.D., 2021. Dopaminergic
Frye, M.A., Keck, P.E., Leverich, G.S., Rush, A.J., Suppes, T., Pollio, C., Drexhage, H. organization of striatum is linked to cortical activity and brain expression of genes
A., 2002. High rate of autoimmune thyroiditis in bipolar disorder: lack of association associated with psychiatric illness. Sci. Adv. 7 https://doi.org/10.1126/sciadv.
with lithium exposure. Biol. Psychiatry 51, 305–311. https://doi.org/10.1016/ abg1512.
S0006-3223(01)01217-3. McLaren, M.E., Nissim, N.R., Woods, A.J., 2018. The effects of medication use in
Law, A.J., Weickert, C.S., Hyde, T.M., Kleinman, J.E., Harrison, P.J., 2004. Reduced transcranial direct current stimulation: A brief review. Brain Stimulation 11, 52–58.
spinophilin but not microtubule-associated protein 2 expression in the hippocampal https://doi.org/10.1016/j.brs.2017.10.006.
formation in schizophrenia and mood disorders: molecular evidence for a pathology Melo, L., Mosayebi-Samani, M., Ghanavati, E., Nitsche, M.A., Kuo, M.F., 2021. Dosage-
of dendritic spines. Am. J. Psychiatry 161, 1848–1855. https://doi.org/10.1176/ dependent impact of acute serotonin enhancement on transcranial direct current
ajp.161.10.1848. stimulation effects. Int. J. Neuropsychopharmacol. 24, 787–797. https://doi.org/
Leffa, D.T., de Souza, A., Scarabelot, V.L., Medeiros, L.F., de Oliveira, C., Grevet, E.H., 10.1093/ijnp/pyab035.
Caumo, W., de Souza, D.O., Rohde, L.A.P., Torres, I.L.S., 2016. Transcranial direct Merikangas, K.R., Jin, R., He, J.-P., Kessler, R.C., Lee, S., Sampson, N.A., Viana, M.C.,
current stimulation improves short-term memory in an animal model of attention- Andrade, L.H., Hu, C., Karam, E.G., Ladea, M., Medina-Mora, M.E., Ono, Y., Posada-
deficit/hyperactivity disorder. Eur. Neuropsychopharmacol. 26, 368–377. https:// Villa, J., Sagar, R., Wells, J.E., Zarkov, Z., 2011. Prevalence and correlates of bipolar
doi.org/10.1016/j.euroneuro.2015.11.012. spectrum disorder in the world mental health survey initiative. Arch. Gen. Psychiatry
Li, M.S., Du, X.D., Chu, H.C., Liao, Y.Y., Pan, W., Li, Z., Hung, G.C., 2019. Delayed effect 68, 241–251. https://doi.org/10.1001/archgenpsychiatry.2011.12.
of bifrontal transcranial direct current stimulation in patients with treatment- Meyer, D., Bonhoeffer, T., Scheuss, V., 2014. Balance and stability of synaptic structures
resistant depression: a pilot study. BMC Psychiatry 19, 180. https://doi.org/ during synaptic plasticity. Neuron 82, 430–443. https://doi.org/10.1016/j.
10.1186/s12888-019-2119-2. neuron.2014.02.031.
Lin, L.F., Doherty, D.H., Lile, J.D., Bektesh, S., Collins, F., 1993. GDNF: a glial cell line- Meyer, B., Mann, C., Götz, M., Gerlicher, A., Saase, V., Yuen, K.S.L., Aedo-Jury, F.,
derived neurotrophic factor for midbrain dopaminergic neurons. Science (New York, Gonzalez-Escamilla, G., Stroh, A., Kalisch, R., 2019. Increased Neural Activity in
N.Y.) 260, 1130–1132. https://doi.org/10.1126/science.8493557. Mesostriatal Regions after Prefrontal Transcranial Direct Current Stimulation and l-
Lin, Y.Y., Chang, C.C., Huang, C.C., Tzeng, N.S., Kao, Y.C., Chang, H.A., 2021. Efficacy DOPA Administration. J. Neurosci. 39, 5326–5335. https://doi.org/10.1523/
and neurophysiological predictors of treatment response of adjunct bifrontal jneurosci.3128-18.2019.
transcranial direct current stimulation (tDCS) in treating unipolar and bipolar Mezger, E., Rauchmann, B.S., Brunoni, A.R., Bulubas, L., Thielscher, A., Werle, J.,
depression. J. Affect. Disord. 280, 295–304. https://doi.org/10.1016/j. Mortazavi, M., Karali, T., Stöcklein, S., Ertl-Wagner, B., Goerigk, S., Padberg, F.,
jad.2020.11.030. Keeser, D., 2021. Effects of bifrontal transcranial direct current stimulation on brain
Longo, V., Barbati, S.A., Re, A., Paciello, F., Bolla, M., Rinaudo, M., Miraglia, F., Alù, F., glutamate levels and resting state connectivity: multimodal MRI data for the
Di Donna, M.G., Vecchio, F., Rossini, P.M., Podda, M.V., Grassi, C., 2022.

17
G. D'Urso et al. Progress in Neuropsychopharmacology & Biological Psychiatry 121 (2023) 110672

cathodal stimulation site. Eur. Arch. Psychiatry Clin. Neurosci. 271, 111–122. serotonin transporter binding in depressed patients with bipolar disorder using
https://doi.org/10.1007/s00406-020-01177-0. positron emission tomography. Arch. Gen. Psychiatry 64, 201–208. https://doi.org/
Miklowitz, D.J., Otto, M.W., Frank, E., Reilly-Harrington, N.A., Wisniewski, S.R., 10.1001/archpsyc.64.2.201.
Kogan, J.N., Nierenberg, A.A., Calabrese, J.R., Marangell, L.B., Gyulai, L., Araga, M., Page, M.J., McKenzie, J.E., Bossuyt, P.M., Boutron, I., Hoffmann, T.C., Mulrow, C.D.,
Gonzalez, J.M., Shirley, E.R., Thase, M.E., Sachs, G.S., 2007. Psychosocial treatments Shamseer, L., Tetzlaff, J.M., Akl, E.A., Brennan, S.E., Chou, R., Glanville, J.,
for bipolar depression: a 1-year randomized trial from the Systematic Treatment Grimshaw, J.M., Hróbjartsson, A., Lalu, M.M., Li, T., Loder, E.W., Mayo-Wilson, E.,
Enhancement Program. Arch. Gen. Psychiatry 64, 419–426. https://doi.org/ McDonald, S., McGuinness, L.A., Stewart, L.A., Thomas, J., Tricco, A.C., Welch, V.A.,
10.1001/archpsyc.64.4.419. Whiting, P., Moher, D., 2021. The PRISMA 2020 statement: an updated guideline for
Miller, N.D., Kelsoe, J.R., 2017. Unraveling the biology of bipolar disorder using induced reporting systematic reviews, 372, p. n71. https://doi.org/10.1136/bmj.n71/JBMJ.
pluripotent stem-derived neurons. Bipolar Disord. 19, 544–551. https://doi.org/ Palm, U., Schiller, C., Fintescu, Z., Obermeier, M., Keeser, D., Reisinger, E., Pogarell, O.,
10.1111/bdi.12535. Nitsche, M.A., Möller, H.J., Padberg, F., 2012. Transcranial direct current
Minichino, A., Bersani, F.S., Spagnoli, F., Corrado, A., De Michele, F., Calò, W.K., stimulation in treatment resistant depression: a randomized double-blind, placebo-
Primavera, M., Yang, B., Bernabei, L., Macrì, F., Vergnani, L., Biondi, M., Delle controlled study. Brain Stimulation 5, 242–251. https://doi.org/10.1016/j.
Chiaie, R., 2014. Prefronto-cerebellar transcranial direct current stimulation brs.2011.08.005.
improves sleep quality in euthymic bipolar patients: a brief report. Behav. Neurol. Palm, U., Keeser, D., Hasan, A., Kupka, M.J., Blautzik, J., Sarubin, N., Kaymakanova, F.,
2014, 876521. https://doi.org/10.1155/2014/876521. Unger, I., Falkai, P., Meindl, T., Ertl-Wagner, B., Padberg, F., 2016. Prefrontal
Mishima, T., Nagai, T., Yahagi, K., Akther, S., Oe, Y., Monai, H., Kohsaka, S., Hirase, H., transcranial direct current stimulation for treatment of schizophrenia with
2019. Transcranial direct current stimulation (tDCS) induces adrenergic receptor- predominant negative symptoms: a double-blind, sham-controlled proof-of-concept
dependent microglial morphological changes in mice. eNeuro 6. https://doi.org/ study. Schizophr. Bull. 42, 1253–1261. https://doi.org/10.1093/schbul/sbw041.
10.1523/eneuro.0204-19.2019. Pearlson, G.D., Wong, D.F., Tune, L.E., Ross, C.A., Chase, G.A., Links, J.M., Dannals, R.F.,
Moffa, A.H., Martin, D., Alonzo, A., Bennabi, D., Blumberger, D.M., Benseñor, I.M., Wilson, A.A., Ravert, H.T., Wagner Jr., H.N., et al., 1995. In vivo D2 dopamine
Daskalakis, Z., Fregni, F., Haffen, E., Lisanby, S.H., Padberg, F., Palm, U., Razza, L.B., receptor density in psychotic and nonpsychotic patients with bipolar disorder. Arch.
Sampaio-Jr, B., Loo, C., Brunoni, A.R., 2020. Efficacy and acceptability of Gen. Psychiatry 52, 471–477. https://doi.org/10.1001/
transcranial direct current stimulation (tDCS) for major depressive disorder: An archpsyc.1995.03950180057008.
individual patient data meta-analysis. Prog. Neuro-Psychopharmacol. Biol. Pereira Junior Bde, S., Tortella, G., Lafer, B., Nunes, P., Benseñor, I.M., Lotufo, P.A.,
Psychiatry 99, 109836. https://doi.org/10.1016/j.pnpbp.2019.109836. Machado-Vieira, R., Brunoni, A.R., 2015. The bipolar depression electrical treatment
Monai, H., Hirase, H., 2018. Astrocytes as a target of transcranial direct current trial (BETTER): design, rationale, and objectives of a randomized, sham-controlled
stimulation (tDCS) to treat depression. Neurosci. Res. 126, 15–21. https://doi.org/ trial and data from the pilot study phase. Neural Plasticity 2015, 684025. https://
10.1016/j.neures.2017.08.012. doi.org/10.1155/2015/684025.
Monai, H., Ohkura, M., Tanaka, M., Oe, Y., Konno, A., Hirai, H., Mikoshiba, K., Phillips, M.L., Kupfer, D.J., 2013. Bipolar disorder diagnosis: challenges and future
Itohara, S., Nakai, J., Iwai, Y., Hirase, H., 2016. Calcium imaging reveals glial directions. Lancet (London, England) 381, 1663–1671. https://doi.org/10.1016/
involvement in transcranial direct current stimulation-induced plasticity in mouse s0140-6736(13)60989-7.
brain. Nat. Commun. 7, 11100. https://doi.org/10.1038/ncomms11100. Popova, V., Daly, E.J., Trivedi, M., Cooper, K., Lane, R., Lim, P., Mazzucco, C., Hough, D.,
Mondino, M., Haesebaert, F., Poulet, E., Suaud-Chagny, M.F., Brunelin, J., 2015. Fronto- Thase, M.E., Shelton, R.C., Molero, P., Vieta, E., Bajbouj, M., Manji, H., Drevets, W.
temporal transcranial Direct Current Stimulation (tDCS) reduces source-monitoring C., Singh, J.B., 2019. Efficacy and safety of flexibly dosed esketamine nasal spray
deficits and auditory hallucinations in patients with schizophrenia. Schizophr. Res. combined with a newly initiated oral antidepressant in treatment-resistant
161, 515–516. https://doi.org/10.1016/j.schres.2014.10.054. depression: a randomized double-blind active-controlled study. Am. J. Psychiatry
Monte-Silva, K., Kuo, M.F., Liebetanz, D., Paulus, W., Nitsche, M.A., 2010a. Shaping the 176, 428–438. https://doi.org/10.1176/appi.ajp.2019.19020172.
optimal repetition interval for cathodal transcranial direct current stimulation Prowacki, M., Lim, L.W., Aquili, L., 2022. Eyeblink rate, a putative dopamine marker,
(tDCS). J. Neurophysiol. 103, 1735–1740. https://doi.org/10.1152/jn.00924.2009. predicts negative reinforcement learning by tDCS of the dlPFC. Brain Stimulation 15,
Monte-Silva, K., Liebetanz, D., Grundey, J., Paulus, W., Nitsche, M.A., 2010b. Dosage- 533–535. https://doi.org/10.1016/j.brs.2022.02.009.
dependent non-linear effect of L-dopa on human motor cortex plasticity. J. Physiol. Rahman, A., Reato, D., Arlotti, M., Gasca, F., Datta, A., Parra, L.C., Bikson, M., 2013.
588, 3415–3424. https://doi.org/10.1113/jphysiol.2010.190181. Cellular effects of acute direct current stimulation: somatic and synaptic terminal
Montgomery, S.A., Asberg, M., 1979. A new depression scale designed to be sensitive to effects. J. Physiol. 591, 2563–2578. https://doi.org/10.1113/jphysiol.2012.247171.
change. Br. J. Psychiatry 134, 382–389. https://doi.org/10.1192/bjp.134.4.382. Ranieri, F., Podda, M.V., Riccardi, E., Frisullo, G., Dileone, M., Profice, P., Pilato, F., Di
Mutz, J., Vipulananthan, V., Carter, B., Hurlemann, R., Fu, C.H.Y., Young, A.H., 2019. Lazzaro, V., Grassi, C., 2012. Modulation of LTP at rat hippocampal CA3-CA1
Comparative efficacy and acceptability of non-surgical brain stimulation for the synapses by direct current stimulation. J. Neurophysiol. 107, 1868–1880. https://
acute treatment of major depressive episodes in adults: systematic review and doi.org/10.1152/jn.00319.2011.
network meta-analysis. BMJ 364, l1079. https://doi.org/10.1136/bmj.l1079. Razza, L.B., De Smet, S., Moffa, A., Sudbrack-Oliveira, P., Vanderhasselt, M.-A.,
Nanou, E., Catterall, W.A., 2018. Calcium channels, synaptic plasticity, and Brunoni, A.R., 2021. Follow-up effects of transcranial direct current stimulation
neuropsychiatric disease. Neuron 98, 466–481. https://doi.org/10.1016/j. (tDCS) for the major depressive episode: A systematic review and meta-analysis.
neuron.2018.03.017. Psychiatry Res. 302, 114024 https://doi.org/10.1016/j.psychres.2021.114024.
Newstead, S., Young, H., Benton, D., Jiga-Boy, G., Sienz, M.L.A., Clement, R.M., Boy, F., Rege, S., Hodgkinson, S.J., 2013. Immune dysregulation and autoimmunity in bipolar
2017. Acute and repetitive fronto-cerebellar tDCS stimulation improves mood in disorder: Synthesis of the evidence and its clinical application. Austr. New Zeal. J.
non-depressed participants. Exp. Brain Res. 236, 83–97. https://doi.org/10.1007/ Psychiatry 47, 1136–1151. https://doi.org/10.1177/0004867413499077.
s00221-017-5109-y. Rodríguez-Cano, E., Alonso-Lana, S., Sarró, S., Fernández-Corcuera, P., Goikolea, J.M.,
Nieratschker, V., Kiefer, C., Giel, K., Krüger, R., Plewnia, C., 2015. The COMT Val/Met Vieta, E., Maristany, T., Salvador, R., McKenna, P.J., Pomarol-Clotet, E., 2017.
Polymorphism Modulates Effects of tDCS on Response Inhibition. Brain Stimulation Differential failure to deactivate the default mode network in unipolar and bipolar
8, 283–288. https://doi.org/10.1016/j.brs.2014.11.009. depression. Bipolar Disord. 19, 386–395. https://doi.org/10.1111/bdi.12517.
Nitsche, M.A., Paulus, W., 2000. Excitability changes induced in the human motor cortex Rohan, J.G., Carhuatanta, K.A., McInturf, S.M., Miklasevich, M.K., Jankord, R., 2015.
by weak transcranial direct current stimulation. J. Physiol. 527 (Pt 3), 633–639. Modulating Hippocampal Plasticity with In Vivo Brain Stimulation. J. Neurosci. 35,
https://doi.org/10.1111/j.1469-7793.2000.t01-1-00633.x. 12824–12832. https://doi.org/10.1523/JNEUROSCI.2376-15.2015.
Nitsche, M.A., Paulus, W., 2001. Sustained excitability elevations induced by transcranial Rosenblat, J.D., McIntyre, R.S., 2017. Bipolar disorder and immune dysfunction:
DC motor cortex stimulation in humans. Neurology 57, 1899–1901. https://doi.org/ epidemiological findings, proposed pathophysiology and clinical implications. Brain
10.1212/wnl.57.10.1899. Sci. 7 https://doi.org/10.3390/brainsci7110144.
Nitsche, M.A., Fricke, K., Henschke, U., Schlitterlau, A., Liebetanz, D., Lang, N., Rroji, O., van Kuyck, K., Nuttin, B., Wenderoth, N., 2015. Anodal tDCS over the primary
Henning, S., Tergau, F., Paulus, W., 2003. Pharmacological modulation of cortical motor cortex facilitates long-term memory formation reflecting use-dependent
excitability shifts induced by transcranial direct current stimulation in humans. plasticity. PLoS One 10, e0127270. https://doi.org/10.1371/journal.pone.0127270.
J. Physiol. 553, 293–301. https://doi.org/10.1113/jphysiol.2003.049916. Rush, A.J., Trivedi, M.H., Ibrahim, H.M., Carmody, T.J., Arnow, B., Klein, D.N.,
Nitsche, M.A., Jaussi, W., Liebetanz, D., Lang, N., Tergau, F., Paulus, W., 2004a. Markowitz, J.C., Ninan, P.T., Kornstein, S., Manber, R., Thase, M.E., Kocsis, J.H.,
Consolidation of Human Motor Cortical Neuroplasticity by D-Cycloserine. Keller, M.B., 2003. The 16-Item Quick Inventory of Depressive Symptomatology
Neuropsychopharmacology 29, 1573–1578. https://doi.org/10.1038/sj. (QIDS), clinician rating (QIDS-C), and self-report (QIDS-SR): a psychometric
npp.1300517. evaluation in patients with chronic major depression. Biol. Psychiatry 54 (5),
Nitsche, M.A., Liebetanz, D., Schlitterlau, A., Henschke, U., Fricke, K., Frommann, K., 573–583. https://doi.org/10.1016/s0006-3223(02)01866-8 (Erratum in: Biol
Lang, N., Henning, S., Paulus, W., Tergau, F., 2004b. GABAergic modulation of DC Psychiatry. 2003 Sep 1;54(5):585. PMID: 12946886).
stimulation-induced motor cortex excitability shifts in humans. Eur. J. Neurosci. 19, Sadleir, R.J., Vannorsdall, T.D., Schretlen, D.J., Gordon, B., 2010. Transcranial direct
2720–2726. https://doi.org/10.1111/j.0953-816X.2004.03398.x. current stimulation (tDCS) in a realistic head model. NeuroImage 51, 1310–1318.
Nitsche, M.A., Cohen, L.G., Wassermann, E.M., Priori, A., Lang, N., Antal, A., Paulus, W., https://doi.org/10.1016/j.neuroimage.2010.03.052.
Hummel, F., Boggio, P.S., Fregni, F., Pascual-Leone, A., 2008. Transcranial direct Salvoro, C., Bortoluzzi, S., Coppe, A., Valle, G., Feltrin, E., Mostacciuolo, M.L., Vazza, G.,
current stimulation: State of the art 2008. Brain Stimulation 1, 206–223. https://doi. 2018. Rare risk variants identification by identity-by-descent mapping and whole-
org/10.1016/j.brs.2008.06.004. exome sequencing implicates neuronal development pathways in schizophrenia and
Nitsche, M.A., Kuo, M.F., Karrasch, R., Wächter, B., Liebetanz, D., Paulus, W., 2009. bipolar disorder. Mol. Neurobiol. 55, 7366–7376. https://doi.org/10.1007/s12035-
Serotonin affects transcranial direct current-induced neuroplasticity in humans. Biol. 018-0922-2.
Psychiatry 66, 503–508. https://doi.org/10.1016/j.biopsych.2009.03.022. Sampaio-Junior, B., Tortella, G., Borrione, L., Moffa, A.H., Machado-Vieira, R., Cretaz, E.,
Oquendo, M.A., Hastings, R.S., Huang, Y.Y., Simpson, N., Ogden, R.T., Hu, X.Z., Fernandes da Silva, A., Fraguas, R., Aparício, L.V., Klein, I., Lafer, B., Goerigk, S.,
Goldman, D., Arango, V., Van Heertum, R.L., Mann, J.J., Parsey, R.V., 2007. Brain Benseñor, I.M., Lotufo, P.A., Gattaz, W.F., Brunoni, A.R., 2018. Efficacy and safety of

18
G. D'Urso et al. Progress in Neuropsychopharmacology & Biological Psychiatry 121 (2023) 110672

transcranial direct current stimulation as an add-on treatment for bipolar depression: extracellular dopamine levels in the rat striatum. Front. Syst. Neurosci. 7, 6. https://
a randomized clinical trial. JAMA Psychiatry 75, 158–166. https://doi.org/10.1001/ doi.org/10.3389/fnsys.2013.00006.
jamapsychiatry.2017.4040. Tiraboschi, E., Giambelli, R., D'Urso, G., Galietta, A., Barbon, A., de Bartolomeis, A.,
Santana-Gómez, C.E., Alcántara-González, D., Luna-Munguia, H., Bañuelos-Cabrera, I., Gennarelli, M., Barlati, S., Racagni, G., Popoli, M., 2004. Antidepressants activate
Magdaleno-Madrigal, V., Tamayo, M., Rocha, L.L., Besio, W.G., 2015. Transcranial CaMKII in neuron cell body by Thr286 phosphorylation. Neuroreport 15,
focal electrical stimulation reduces seizure activity and hippocampal glutamate 2393–2396. https://doi.org/10.1097/00001756-200410250-00018.
release during status epilepticus. In: 2015 37th Annual International Conference of Tomasetti, C., Dell'Aversano, C., Iasevoli, F., de Bartolomeis, A., 2007. Homer splice
the IEEE Engineering in Medicine and Biology Society (EMBC), pp. 6586–6589. variants modulation within cortico-subcortical regions by dopamine D2 antagonists,
Sauvaget, A., Poulet, E., Mantovani, A., Bulteau, S., Damier, P., Moutaud, B., a partial agonist, and an indirect agonist: implication for glutamatergic postsynaptic
Paternoster, M., de Bartolomeis, A., D'Urso, G., 2018. The Psychiatric density in antipsychotics action. Neuroscience 150, 144–158. https://doi.org/
Neuromodulation Unit: Implementation and Management. J. ECT 34, 211–219. 10.1016/j.neuroscience.2007.08.022.
https://doi.org/10.1097/yct.0000000000000513. Tomasetti, C., Dell'Aversano, C., Iasevoli, F., Marmo, F., de Bartolomeis, A., 2011. The
Sauvaget, A., Tostivint, A., Etcheverrigaray, F., Pichot, A., Dert, C., Schirr-Bonnais, S., acute and chronic effects of combined antipsychotic-mood stabilizing treatment on
Clouet, J., Sellal, O., Mauduit, N., Leux, C., Cabelguen, C., Bulteau, S., Riche, V.P., the expression of cortical and striatal postsynaptic density genes. Prog. Neuro-
2019. Hospital production cost of transcranial direct current stimulation (tDCS) in Psychopharmacol. Biol. Psychiatry 35, 184–197. https://doi.org/10.1016/j.
the treatment of depression. Neurophysiologie clinique =. Clin. Neurophysiol. 49, pnpbp.2010.10.025.
11–18. https://doi.org/10.1016/j.neucli.2018.11.001. Tomasetti, C., Iasevoli, F., Buonaguro, E.F., De Berardis, D., Fornaro, M., Fiengo, A.L.,
Schröter, K., Brum, M., Brunkhorst-Kanaan, N., Tole, F., Ziegler, C., Domschke, K., Martinotti, G., Orsolini, L., Valchera, A., Di Giannantonio, M., de Bartolomeis, A.,
Reif, A., Kittel-Schneider, S., 2020. Longitudinal multi-level biomarker analysis of 2017. Treating the synapse in major psychiatric disorders: the role of postsynaptic
BDNF in major depression and bipolar disorder. Eur. Arch. Psychiatry Clin. Neurosci. density network in dopamine-glutamate interplay and psychopharmacologic drugs
270, 169–181. https://doi.org/10.1007/s00406-019-01007-y. molecular actions. Int. J. Mol. Sci. 18 https://doi.org/10.3390/ijms18010135.
Serchov, T., Clement, H.-W., Schwarz, Martin K., Iasevoli, F., Tosh, Dilip K., Idzko, M., Tylee, D.S., Sun, J., Hess, J.L., Tahir, M.A., Sharma, E., Malik, R., Worrall, B.B., Levine, A.
Jacobson, Kenneth A., de Bartolomeis, A., Normann, C., Biber, K., van Calker, D., J., Martinson, J.J., Nejentsev, S., Speed, D., Fischer, A., Mick, E., Walker, B.R.,
2015. Increased Signaling via Adenosine A1 Receptors, Sleep Deprivation, Crawford, A., Grant, S.F.A., Polychronakos, C., Bradfield, J.P., Sleiman, P.M.A.,
Imipramine, and Ketamine Inhibit Depressive-like Behavior via Induction of Hakonarson, H., Ellinghaus, E., Elder, J.T., Tsoi, L.C., Trembath, R.C., Barker, J.N.,
Homer1a. Neuron 87, 549–562. https://doi.org/10.1016/j.neuron.2015.07.010. Franke, A., Dehghan, A., Faraone, S.V., Glatt, S.J., 2018. Genetic correlations among
Shin, D.W., Fan, J., Luu, E., Khalid, W., Xia, Y., Khadka, N., Bikson, M., Fu, B.M., 2020. In psychiatric and immune-related phenotypes based on genome-wide association data.
Vivo Modulation of the Blood-Brain Barrier Permeability by Transcranial Direct Am. J. Med. Genet. B Neuropsychiatric Genet. 177, 641–657. https://doi.org/
Current Stimulation (tDCS). Ann. Biomed. Eng. 48, 1256–1270. https://doi.org/ 10.1002/ajmg.b.32652.
10.1007/s10439-020-02447-7. Valvassori, S.S., Mariot, E., Varela, R.B., Bavaresco, D.V., Dal-Pont, G.C., Ferreira, C.L.,
Sidor, M.M., MacQueen, G.M., 2012. An update on antidepressant use in bipolar Andersen, M.L., Tye, S.J., Quevedo, J., 2019. The role of neurotrophic factors in
depression. Curr. Psychiatry Rep. 14, 696–704. https://doi.org/10.1007/s11920- manic-, anxious- and depressive-like behaviors induced by amphetamine
012-0323-6. sensitization: Implications to the animal model of bipolar disorder. J. Affect. Disord.
Soares-Weiser, K., Bravo Vergel, Y., Beynon, S., Dunn, G., Barbieri, M., Duffy, S., 245, 1106–1113. https://doi.org/10.1016/j.jad.2018.10.370.
Geddes, J., Gilbody, S., Palmer, S., Woolacott, N., 2007. A systematic review and van Enkhuizen, J., Geyer, M.A., Minassian, A., Perry, W., Henry, B.L., Young, J.W., 2015.
economic model of the clinical effectiveness and cost-effectiveness of interventions Investigating the underlying mechanisms of aberrant behaviors in bipolar disorder
for preventing relapse in people with bipolar disorder. Health Technol. Assess. 11 from patients to models: Rodent and human studies. Neurosci. Biobehav. Rev. 58,
https://doi.org/10.3310/hta11390 iii-iv, ix-206. 4–18. https://doi.org/10.1016/j.neubiorev.2015.08.008.
Sommerlandt, F.M.J., Brockmann, A., Rössler, W., Spaethe, J., 2019. Immediate early Veenakumari, M., Goyal, N., Kumar, M., Kshitiz, K.K., Kumar, P., 2022. Serum glial cell
genes in social insects: a tool to identify brain regions involved in complex behaviors derived neurotrophic factor (GDNF) as a predictor of response to HD-tDCS in bipolar
and molecular processes underlying neuroplasticity. Cell. Mol. Life Sci. 76, 637–651. affective disorder. Asian J. Psychiatr. 68, 102965 https://doi.org/10.1016/j.
https://doi.org/10.1007/s00018-018-2948-z. ajp.2021.102965.
Stagg, C.J., Best, J.G., Stephenson, M.C., O’Shea, J., Wylezinska, M., Kincses, Z.T., Vieta, E., Locklear, J., Günther, O., Ekman, M., Miltenburger, C., Chatterton, M.L.,
Johansen-Berg, H., 2009. Polarity-sensitive modulation of cortical neurotransmitters Aström, M., Paulsson, B., 2010. Treatment options for bipolar depression: a
by transcranial stimulation. J. Neurosci. 29 (16), 5202–5206. https://doi.org/ systematic review of randomized, controlled trials. J. Clin. Psychopharmacol. 30,
10.1523/jneurosci.4432-08.2009. 579–590. https://doi.org/10.1097/JCP.0b013e3181f15849.
Sun, P., Wang, F., Wang, L., Zhang, Y., Yamamoto, R., Sugai, T., Zhang, Q., Wang, Z., Wong, D.F., Pearlson, G.D., Tune, L.E., Young, L.T., Meltzer, C.C., Dannals, R.F.,
Kato, N., 2011. Increase in cortical pyramidal cell excitability accompanies Ravert, H.T., Reith, J., Kuhar, M.J., Gjedde, A., 1997. Quantification of
depression-like behavior in mice: a transcranial magnetic stimulation study. neuroreceptors in the living human brain: IV. Effect of aging and elevations of D2-
J. Neurosci. 31, 16464–16472. https://doi.org/10.1523/jneurosci.1542-11.2011. like receptors in schizophrenia and bipolar illness. J. Cereb. Blood Flow Metab 17,
Suppes, T., Kelly, D.I., Perla, J.M., 2005. Challenges in the management of bipolar 331–342. https://doi.org/10.1097/00004647-199703000-00010.
depression. J. Clin. Psychiatry 66 (Suppl. 5), 11–16. Xia, Y., Khalid, W., Yin, Z., Huang, G., Bikson, M., Fu, B.M., 2020. Modulation of solute
Takano, Y., Yokawa, T., Masuda, A., Niimi, J., Tanaka, S., Hironaka, N., 2011. A rat diffusivity in brain tissue as a novel mechanism of transcranial direct current
model for measuring the effectiveness of transcranial direct current stimulation stimulation (tDCS). Sci. Rep. 10, 18488. https://doi.org/10.1038/s41598-020-
using fMRI. Neurosci. Lett. 491, 40–43. https://doi.org/10.1016/j. 75460-4.
neulet.2011.01.004. Yu, T.-H., Wu, Y.-J., Chien, M.-E., Hsu, K.-S., 2019. Transcranial direct current
Tanaka, T., Takano, Y., Tanaka, S., Hironaka, N., Kobayashi, K., Hanakawa, T., stimulation induces hippocampal metaplasticity mediated by brain-derived
Watanabe, K., Honda, M., 2013. Transcranial direct-current stimulation increases neurotrophic factor. Neuropharmacology 144, 358–367. https://doi.org/10.1016/j.
neuropharm.2018.11.012.

19

You might also like