ArticlePDF Available

Revisiting the antidepressant-like effects of desipramine in male and female adult rats: sex disparities in neurochemical correlates

Authors:

Abstract and Figures

Background The preclinical antidepressant-like characterization of desipramine relied almost exclusively in male rodents, with only a few contradictory reports done in females. Given that most experiments assessed a single dose and/or timepoint of analysis after-treatment, this study evaluated potential sex-differences in the length of the antidepressant-like response induced by different doses of desipramine as well as the molecular underpinnings driving the different responses by sex. Methods Male and female Sprague–Dawley rats were treated (i.p.) with 3 pulses of desipramine (5, 10 or 20 mg/kg) or vehicle (0.9% NaCl) within 24 h. The antidepressant-like effects were evaluated in the forced-swim test 1-h, 1- and 3-day post-treatment. The rate of cell proliferation and the regulation of key neuroplasticity markers (FADD, Cdk5, p35, p25) involved in antidepressant-like responses in the hippocampus were evaluated 1-h, 1-day and 5-day post-treatment. Results Desipramine induced similar antidepressant-like effects in male and female rats (effective doses of 10 and 20 mg/kg, with effects that lasted up to 1-day post-treatment), without altering the rate of cell proliferation. However, some sex-differences emerged when evaluating neuroplasticity markers in the hippocampus, while no changes were observed for female rats, desipramine regulated FADD, Cdk-5 and p25 in males in a way that suggested neuroprotective actions. Conclusions Our findings imply that while desipramine induced similar antidepressant-like responses for male and female rats, some differences emerged in the regulation of certain neuroplasticity markers, suggesting that distinctive molecular mechanisms might be participating in the therapeutic response of desipramine for both sexes.
Content may be subject to copyright.
Vol:.(1234567890)
Pharmacological Reports (2022) 74:626–636
https://doi.org/10.1007/s43440-022-00372-1
1 3
ARTICLE
Revisiting theantidepressant‑like effects ofdesipramine inmale
andfemale adult rats: sex disparities inneurochemical correlates
SandraLedesma‑Corvi1,2· M.JuliaGarcía‑Fuster1,2
Received: 4 April 2022 / Revised: 12 May 2022 / Accepted: 13 May 2022 / Published online: 2 June 2022
© The Author(s) 2022
Abstract
Background The preclinical antidepressant-like characterization of desipramine relied almost exclusively in male rodents,
with only a few contradictory reports done in females. Given that most experiments assessed a single dose and/or timepoint
of analysis after-treatment, this study evaluated potential sex-differences in the length of the antidepressant-like response
induced by different doses of desipramine as well as the molecular underpinnings driving the different responses by sex.
Methods Male and female Sprague–Dawley rats were treated (i.p.) with 3 pulses of desipramine (5, 10 or 20mg/kg) or
vehicle (0.9% NaCl) within 24h. The antidepressant-like effects were evaluated in the forced-swim test 1-h, 1- and 3-day
post-treatment. The rate of cell proliferation and the regulation of key neuroplasticity markers (FADD, Cdk5, p35, p25)
involved in antidepressant-like responses in the hippocampus were evaluated 1-h, 1-day and 5-day post-treatment.
Results Desipramine induced similar antidepressant-like effects in male and female rats (effective doses of 10 and 20mg/
kg, with effects that lasted up to 1-day post-treatment), without altering the rate of cell proliferation. However, some sex-
differences emerged when evaluating neuroplasticity markers in the hippocampus, while no changes were observed for female
rats, desipramine regulated FADD, Cdk-5 and p25 in males in a way that suggested neuroprotective actions.
Conclusions Our findings imply that while desipramine induced similar antidepressant-like responses for male and female
rats, some differences emerged in the regulation of certain neuroplasticity markers, suggesting that distinctive molecular
mechanisms might be participating in the therapeutic response of desipramine for both sexes.
Keywords Antidepressant· Rat· Hippocampus· Neuroplasticity· Sex
Introduction
There are well-known sex-differences in depression rates,
with women being almost twice as likely to suffer depres-
sion as men (e.g., [13]). Moreover, an extensive body of
research demonstrated sex differences in antidepressant
efficacy (reviewed by [4, 5]), although the direction of the
change is not completely clear; while generally speaking
women seem less responsive and/or suffering more adverse
effects than men, this appears especially true for tricyclic
antidepressants; however, other reports suggested a better
efficacy for females might also exist for drugs selectively
inhibiting the serotonin reuptake (reviewed by [6]). These
sex differences in efficacy might be explained by disparities
in the pharmacokinetics and, to a lesser extent, pharmacody-
namics effects of the drug (reviewed by [47]), which com-
bined with the shortage of preclinical studies performed in
female rats [8], justify the poor translation to the clinic, spe-
cially for female patients. Adjusting doses and/or timings,
and/or even changing medications should be considered for
each sex for an optimal clinical response.
In an effort to improve the scarce number of studies
including sex as a biological variable [810], our research
group (among several others) is centered in characteriz-
ing sex differences in antidepressant-like responses (e.g.,
[1113]), including revisiting the antidepressant-like effects
of desipramine in male and female rats which was the goal
of the present study. Desipramine, a typical tricyclic anti-
depressant, is an effective and safe treatment prescribed for
major depressive and other psychiatric disorders [14, 15].
Although, desipramine showed no significant sex differences
* M. Julia García-Fuster
j.garcia@uib.es
1 IUNICS, University oftheBalearic Islands, Cra. de
Valldemossa km 7.5, 07122Palma, Spain
2 Health Research Institute oftheBalearic Islands (IdISBa),
Palma, Spain
627
Revisiting theantidepressant‑like effects ofdesipramine inmale andfemale adult rats: sex…
1 3
for the treatment of multiple illnesses in children and ado-
lescents [16], other prior pharmacological studies revealed
an impact of sex in hormonal responses to desipramine
in healthy young sibling pairs in a randomized cross-over
experiment [17], as well as sex differences in pharmacoki-
netics (reviewed by [6]). The preclinical characterization of
desipramine as an antidepressant relied almost exclusively in
male naïve rodents (e.g., [1820]) or male rodents exposed
to stress [21], and generally utilizing the forced-swim test
as a well-accepted experimental tool to screen drugs for
its activity [22]. However, only a few preclinical studies
included females, and showed contradictory results, with
either signs of a significant antidepressant-like response [23]
or a lack of efficacy [24]. In the context of these inconsistent
effects, and given that most studies only assessed a single
dose and/or timepoint of analysis after treatment, the present
study evaluated potential sex differences in the length of
the antidepressant-like response (1h, 1day and up to 5-day
post-treatment) induced by different doses of desipramine (5,
10 and 20mg/kg) in rats exposed to the forced-swim test, a
model with strong predictive and discriminative validity, in
which to further characterize the molecular underpinnings
driving the potential different responses of desipramine by
sex.
In particular, neural adaptations taking place in the hip-
pocampus (reviewed by [25], and more recently by [26]),
such as neurogenesis, are modulated in depressive-like states
(neurogenic theory of depression; [2729]), by most antide-
pressants [28, 30]), desipramine in particular [21, 3133],
and its ablation reduced the effects of certain antidepressants
[34, 35], suggesting an important role for this particular pro-
cess in the antidepressant-like response of certain drugs.
Moreover, besides neurogenesis, other hippocampal plas-
ticity events [25, 26] induced by desipramine also deserve
further characterization, in terms of their role in controlling
cell fate (balance between cell death vs. survival/plasticity),
and since these processes might alter the basal rates of cell
genesis, and participate in mediating some of the behavio-
ral responses observed. For example, since depression has
been linked to certain neurotoxic events (aberrant apopto-
sis, [36]) in humans and animals (reviewed by [25]), the
role of antidepressants as neuroprotective agents (mediating
anti-apoptotic actions) generated continued interest. Particu-
larly, different antidepressant drugs, including desipramine,
decreased pro-apoptotic signals (e.g., [37]) and promoted
anti-apoptotic functions through the regulation of Fas-
associated protein with death domain (FADD), and inter-
estingly FADD levels correlated with the antidepressant-
like response exerted by desipramine [19]. Finally, cyclin
dependent kinase-5 (Cdk5), which is limited to post-mitotic
neurons [39] and is linked to hippocampal neurogenesis by
playing an important role in neuronal migration and hip-
pocampal cell survival [42], also regulates depression-like
behavior [38], and depends of co-factors p35 and p25 [40,
41]. While hippocampal Cdk-5 activity was increased in
animal models of stress, it was reduced by antidepressants
through redistribution of p35 from the cell membrane to the
cytoplasm [43], suggesting that the Cdk-5/p35–p25 complex
might play an important role in regulating antidepressant
actions. Against this background, and since most molecular
studies previously reported were performed almost exclu-
sively in male rodents, this study deepened in the molecular
sex differences induced by desipramine in male and female
rats and developing throughout time.
Experimental procedures
Animals
Adult Sprague–Dawley rats (n = 142; 90 males and 52
females) were bred at the University of the Balearic Islands
and housed in groups of 2–4 in standard cages in controlled
environmental conditions (22ºC, 70% humidity, and 12-h
light/dark cycle, lights on at 8:00 a.m.) with ad libitum
access to a standard diet and tap water. Prior to the experi-
mental procedures, rats were habituated to being handled
by the experimenter (for at least 2days). All animal pro-
cedures were performed during the light period, followed
ARRIVE guidelines [44, 45] and Directive 2010/63/EU of
the European Parliament and of the Council, and were pre-
viously approved by the Local Bioethical Committee (Uni-
versity of the Balearic Islands; CEEA 58/04/16 and CEEA
155/12/20) and the Regional Government (Conselleria Medi
Ambient, Agricultura i Pesca, Direcció General Agricultura
i Ramaderia, Govern de les Illes Balears; Exp.: 2016/08/
AEXP and 2021/05/AEXP) as stated in the Spanish Royal
Decree 53/2013. All efforts were made to minimize the num-
ber of rats used and their suffering. In this context, to avoid
inducing extra-stress in female rats during the experimental
procedure, and since only a minor influence on the baseline
response for naturally cycling females was reported in the
forced-swim test (reviewed by [46]), and the cyclicity of
females was not part of our research question (see [10]), the
specific stages of the estrous cycle were not examined.
Pharmacological treatments
In a series of independent experiments (i.e., female and
male rats were performed at different points in time), ran-
domly allocated rats from each sex received (i.p.), 3 pulses
(23, 5 and 1h prior to behavioral or neurochemical test-
ing; see Fig.1) of desipramine (5, 10 or 20mg/kg; doses
selected from [19]) or saline (0.9% NaCl, 1ml/kg, i.p.)
within a period of 24h, at the indicated times (see Fig.1).
The rats exposed to behavioral testing were sacrificed 5-day
628 S.Ledesma-Corvi, M.J.García-Fuster
1 3
post-treatment (n = 74, see Fig. 1A), while other parallel
experiments were performed in rats from each sex (with no
behavioral testing) to collect brains at different timepoints
after treatment (1-h and 1-day post-treatment; see Fig.1B).
Forced‑swim test
A cohort of rats was used to evaluate the antidepressant-
like potential of desipramine (control, n = 17 males and 8
females; desipramine 5mg/kg, n = 9 males and 7 females;
desipramine 10mg/kg, n = 9 males and 8 females; desip-
ramine 20mg/kg; n = 8 males and 8 females; see Fig.1A).
To do so, we relied on the forced-swim test, with predic-
tive validity for assessing antidepressant-like efficacy [22],
and with prior reliable results from our research group (e.g.,
[19, 47, 48]), including the evaluation of the time-course
length of the response (e.g., [1113]). To perform this test,
we used water tanks of 41cm high × 32cm diameter, filled
with clean water for each rat to a depth of 25cm and at a
temperature of 25 ± 1°C. Prior to any drug treatment, rats
were exposed to a pre-test forced-swim session (15min;
D1, see Fig.1A), followed by a sequence of 5-min tests
performed 1-h, 1-day and 3-day post-treatment to evaluate
duration of the antidepressant-like response of desipramine.
Sessions were videotaped and analyzed by an experimenter
blind to the treatment groups with Behavioral Tracker soft-
ware (CA, USA) that provided a measure of the time rats
spent immobile vs. active (i.e., climbing). Since experi-
ments were done in different waves throughout the year,
and given the described circannual changes in the duration
of the immobility response of rats in the forced-swim test
[49], among other factors also affecting baseline levels in
the forced-swim test (reviewed by [50]), direct comparisons
across studies (in terms of the individual seconds spent in
each behavior) were not applicable and thus, results were
calculated for each study as the percent change vs. the cor-
responding wave-control group for each sex.
Tissue collection
Brain tissue from different cohort of rats was collected at the
different timepoints of analysis (1h, 1 and 5days, see Fig.1)
in which to evaluate neuroplastic-like changes that might
emerge during the course of treatment and that could explain
the behavioral-like responses observed (control, 1h: n = 12
males and 6 females; 1day: n = 11 males and 4 females;
desipramine 10mg/kg, 1h: n = 6 males; 1day: n = 6 males;
desipramine 20mg/kg; 1h: n = 6 males and 6 females; 1day:
Fig. 1 Experimental design. A Time-course of the antidepressant-like
response induced by different doses of desipramine was evaluated in
the forced-swim test (FST) 1-h and 1- and 3-day post-treatment in
male and female rats (control-vehicle, n = 17 males and 8 females;
desipramine 5mg/kg, n = 9 males and 7 females; desipramine 10mg/
kg, n = 9 males and 8 females; desipramine 20mg/kg; n = 8 males and
8 females). Prior to any drug treatment, rats were handled for some
days and then exposed to a 15-min pre-forced-swim test (D1), fol-
lowed by 3 pulses within a 24-h period of the corresponding treat-
ment, and a sequence of 5-min forced-swim tests performed 1-h (D2),
1-day (D3) and 3-day (D5) post-treatment. Brains were collected
5-day post-treatment (D7) to evaluate potential neurochemical mark-
ers regulated by desipramine through immunohistochemistry (IHC)
or western blot (WB) analyses. B Potential neurochemical markers
regulated by desipramine were also evaluated by immunohistochem-
istry (IHC) or western blot (WB) analyses 1-h and 1-day post-treat-
ment in male and female rats treated with the same drug regimens
(control, 1h: n = 12 males and 6 females; 1 day: n = 11 males and 4
females; desipramine 10mg/kg, 1h: n = 6 males; 1 day: n = 6 males;
desipramine 20mg/kg; 1h: n = 6 males and 6 females; 1 day: n = 6
males and 5 females)
629
Revisiting theantidepressant‑like effects ofdesipramine inmale andfemale adult rats: sex…
1 3
n = 6 males and 5 females; see Fig.1). All rats were killed by
rapid decapitation and their brains removed at the indicated
times. The left half-brain was quickly frozen in isopentane
at – 30ºC and stored at – 80ºC, until it was cryostat-cut
in 30μm sections starting at around − 0.72mm and up to
-6.80mm from Bregma, to cover the whole hippocampal
extent (starting around − 1.72 from Bregma). Sections were
slide-mounted and kept at – 80ºC for the later analysis of
the early stages of hippocampal neurogenesis (i.e., Ki-67
for recent cell proliferation) by immunohistochemistry. The
right hippocampus was freshly dissected, frozen in liquid
nitrogen, and kept at – 80ºC until neuroplasticity markers
(i.e., FADD, Cdk-5, p35-p25) were examined by Western
blot experiments.
Immunohistochemistry
The rate of cell proliferation was quantified in tissuesections
(30μm) that were post-fixed in 4% paraformaldehyde and
exposed to several steps (i.e., antigen retrieval, blocking),
including the incubation with the primary antibody anti-
Ki-67 (1:40,000, kindly provided by Professors Huda Akil
and Stanley J. Watson, University of Michigan, MI, USA)
and as previously described (for further details see [51, 52]).
Ki-67 + cells were quantified in the dentate gyrus with a
Leica DMR light microscope (63 × objective lens) in every
8th section throughout the entire extent of the hippocampus
following a modified unbiased stereological procedure [51,
52], and then the overall number of cells was multiplied
by the sampling factor 8 to provide an estimate of the total
number labeled cells per animal (e.g., [11, 51, 52]).
Western blot
Following standardized protocols from our research group
(e.g., [19, 53]), total brain proteins (40μg) from rat hip-
pocampal homogenates were resolved by electrophoresis
on 10–12% SDS–PAGE minigels (Bio-Rad Laboratories,
Hercules, CA, USA) and then transferred to nitrocellulose
membranes that were incubated overnight at 4 ºC in block-
ing solution containing the appropriate primary antibodies
(previously characterized, see [19, 53]). The vendors and
conditions of the primary antibodies were the following: (1)
Santa Cruz Biotechnology (CA, USA): anti-FADD (H-181)
(dilution 1:5000) and anti-p35 (Ab-C19) (dilution 1:3000),
(2) Lab Vision Corporation (CA, USA): anti-Cdk-5 (DC17)
(dilution 1:1000), and (3) Sigma-Aldrich (MO, USA): anti-
β-actin (AC-15) (dilution 1:10,000). Following the incu-
bation (1h) at room temperature with the corresponding
secondary antibody (anti-rabbit or anti-mouse, 1:5000 dilu-
tion; Cell Signaling, MA, USA), target proteins were visual-
ized by exposing membranes incubated with ECL reagents
(Amersham, Buckinghamshire, UK) to an autoradiographic
film (Amersham ECL Hyperfilm) for 1–60min. The signal
was quantified by densitometric scanning (GS-800 Imaging
Calibrated Densitometer, Bio-Rad) and immunoreactivity
of target proteins (i.e., FADD, Cdk-5, p35–p25 and β-actin)
for each rat was calculated in each gel as the percent (%)
change vs. the corresponding control group (1 or 5days).
Each sample was loaded at least 3 times in different gels and
the mean value was used as a final estimate. β-actin served
as a loading control, since its content was not altered by any
treatment conditions (data not shown).
Data analysis anddata availability statement
Data were analyzed with GraphPad Prism, Version 9.3.1
(CA, USA). Bar graphs represent mean values ± standard
error of the mean (SEM), and contain individual symbols
for each rat, in line with the guidelines for displaying data
and statistical methods in experimental pharmacology [54,
55]. Normal distribution was evaluated with Shapiro–Wilk
normality test and thus parametric tests were used for data
evaluation. Behavioral changes in the forced-swim test were
evaluated by two-way repeated measures (RM) ANOVAs
followed by Dunnett’s multiple comparisons tests when
appropriate, in which Treatment (control vs. desipramine:
5, 10 and 20mg/kg) and Time (1-h, 1- and 3-day post-treat-
ment) were used as independent variables. The regulation
of hippocampal cell genesis (Ki-67 + cells) and neuroplas-
ticity markers were evaluated at each timepoint of analysis
for male rats by one-way ANOVAs followed by Dunnett’s
multiple comparison tests when appropriate, and for female
rats by Student's t tests (control vs. 20mg/kg groups). The
variable Time was not included in the neurochemical analy-
sis, since brains were collected at each timepoint of analy-
sis from independent experiments (see Fig.1), and given
the potential basal differences in the regulation of certain
molecular markers with the particular conditions at the time
of sample collecting. The level of significance was set at
p 0.05. The data that supports the findings of this study
will be available upon reasonable request to the correspond-
ing author.
Results
Desipramine induced similar antidepressant‑like
responses inmale andfemale rats
The antidepressant-like effect of desipramine was evaluated
across time (1-h, 1- and 3-day post-treatment) in the forced-
swim test in male rats by two-way RM ANOVAs. The results
showed significant Treatment x Day interactions for immo-
bility (F6,68 = 4.82, p < 0.001) and climbing (F6,68 = 10.78,
p < 0.001; Fig.2A). Dunnett’s post-hoc comparisons revealed
630 S.Ledesma-Corvi, M.J.García-Fuster
1 3
that desipramine decreased immobility in a dose-dependent
manner as observed 1-h (5mg/kg: -8 ± 10%, p = 0.799, n.s.;
10mg/kg: − 39 ± 6%, ***p < 0.001; 20mg/kg: 40 ± 11%,
*p = 0.013) and 1-day post-treatment (5mg/kg: + 10 ± 8%,
p = 0.554, n.s.; 10mg/kg: − 38 ± 11%, *p = 0.011; 20mg/
kg: 35 ± 14%, p = 0.062, n.s.) and as compared with
control-treated rats. Similarly, Dunnett’s post-hoc com-
parisons for climbing showed significant increases 1-h
(5mg/kg: + 8 ± 21%, p = 0.971, n.s.; 10mg/kg: + 55 ± 16%,
*p = 0.010; 20mg/kg: 314 ± 94%, *p = 0.030) and 1-day
post-treatment (5 mg/kg: − 25 ± 20%, p = 0.496, n.s.;
10mg/kg: + 88 ± 28%, *p = 0.017; 20 mg/kg: + 157 ± 52%,
*p = 0.039) and as compared with control-treated rats
(Fig.2A). For female rats, similar results were observed:
Treatment x Day interactions for immobility (F6,54 = 3.49,
p = 0.006) and climbing (F6,54 = 4.32, p = 0.001; Fig.2B) and
significant Dunnett’s post-hoc comparisons for immobility
(1h: 5mg/kg, − 4 ± 5%, p = 0.756, n.s.; 10mg/kg, -26 ± 9%,
*p = 0.037; 20mg/kg, − 50 ± 11%, **p = 0.005; 1day: 5mg/
kg, + 1 ± 4%, p = 0.997, n.s.; 10mg/kg, − 4 ± 6%, p = 0.830,
n.s.; 20mg/kg, − 27 ± 8%, *p = 0.015) and climbing (1h:
5mg/kg, + 33 ± 47%, p = 0.820, n.s.; 10mg/kg, + 244 ± 80%,
*p = 0.035; 20 mg/kg, + 470 ± 106%, **p = 0.005; 1day:
5mg/kg, 0 ± 53%, p > 0.999, n.s.; 10 mg/kg, + 64 ± 79%,
p = 0.767, n.s.; 20mg/kg, + 339 ± 97%, *p = 0.015). All the
changes induced bydesipramine on immobility and climbing
in male and female rats returned to normal 3-day post-treat-
ment (see Fig.2A and 2B).
Desipramine did notregulate therate ofcell
proliferation inthedentate gyrus ofmale
andfemale rats
The effect of desipramine on regulating an early marker of
hippocampal neurogenesis (i.e., cell proliferation by labeling
Ki-67 + cells) was evaluated 1-h and 1-day post-treatment
(timepoints with an observed antidepressant-like effect).
The results showed that desipramine did not significantly
alter the number of Ki-67 + cells observed in the hippocam-
pus of male (1h: F2,20 = 2.67, p = 0.094; 1day: F2,20 = 0.34,
p = 0.715) and female (1h: t = 2.21, df = 10, p = 0.052; 1day:
t = 0.77, df = 7, p = 0.467) rats (see Fig.3).
Desipramine modulated certain neuroplasticity
markers inthehippocampus ofmale butnotfemale
rats
When assessing FADD hippocampal regulation, one-way
ANOVAs detected significant effects 1 h (F2,20 = 4.81,
p = 0.020) and 1day (F2,20 = 18.11, p < 0.001), but not
5days (F2,29 = 0.25, p = 0.779) post-treatment (Fig. 4A).
Dunnett’s multiple comparisons tests revealed that the dose
of 20mg/kg of desipramine decreased FADD content 1-h
Fig. 2 Evaluating the time-course of the antidepressant-like response
of different doses of desipramine in male and female rats. Groups of
treatment (i.p., 3 pulses within a 24-h period): control (C: 0.9% NaCl,
1 ml/kg; n = 17 males and 8 females), desipramine (DMI) 5 mg/kg
(n = 9 males and 7 females), 10 mg/kg (n = 9 males and 8 females),
or 20 mg/kg (n = 8 males and 8 females). Columns represent
mean ± SEM of the % time spent immobile or climbing (individual
symbols are shown for each rat). Two-way RM ANOVAs followed
by Dunnett's multiple comparisons tests: ***p < 0.001, **p < 0.01 or
*p < 0.05 when compared to control rats at the indicated timepoint of
analysis
631
Revisiting theantidepressant‑like effects ofdesipramine inmale andfemale adult rats: sex…
1 3
post-treatment (− 93 ± 33%, *p = 0.022), while increased it
1-day post-treatment (+ 90 ± 16%, ***p < 0.001) as com-
pared to their respective control treated rats (Fig.4A).
No significant effects were observed for female rats (1h:
t = 1.86, df = 10, p = 0.093; 1day: t = 2.03, df = 7, p = 0.082;
5days: t = 0.06, df = 13, p = 0.956; Fig.4A).
As for Cdk5, one-way ANOVAs detected signifi-
cant effects 1-day (F2,20 = 12.72, p < 0.001), but not 1-h
(F2,20 = 1.82, p = 0.189) or 5-day (F2,29 = 0.17, p = 0.848)
post-treatment (Fig.4B). Dunnett’s multiple compari-
sons tests revealed that the dose of 20mg/kg of desipra-
mine increased Cdk5 protein content 1-day post-treatment
(+ 33 ± 7%, *p = 0.022) as compared to their respective
control treated rats (Fig.4B). No significant effects were
observed for female rats (1h: t = 0.07, df = 10, p = 0.948;
1day: t = 0.25, df = 7, p = 0.812; 5 days: t = 2.06, df = 14,
p = 0.059; Fig.4B).
The analysis of hippocampal p35 revealed a significant
effect 1h (F2,20 = 4.05, p = 0.033), but not 1-day (F2,20 = 2.92,
p = 0.077) or 5-day (F2,29 = 2.24, p = 0.125) post-treatment
(Fig.4C). However, no significant pairwise comparisons
were revealed by Dunnett's test 1-h post-treatment. Simi-
larly, to the regulation of the other proteins, no significant
effects were observed for female rats (1h: t = 1.66, df = 10,
p = 0.129; 1day: t = 0.94, df = 7, p = 0.380; 5days: t = 0.31,
df = 13, p = 0.759; Fig.4C).
Finally, when assessing p25 hippocampal regulation, one-
way ANOVAs detected significant effects 1h (F2,20 = 7.70,
p = 0.003) and 1day (F2,20 = 9.47, p = 0.001), but not 5-day
(F2,29 = 2.52, p = 0.098) post-treatment (Fig. 4D). Dun-
nett’s multiple comparisons tests revealed that the dose of
20mg/kg of desipramine decreased p25 content as meas-
ured 1-h (− 43 ± 12%, **p = 0.005) and 1-day (− 31 ± 9%,
**p = 0.003) post-treatment, and as compared to their
respective control groups (Fig.4C). Again, no significant
effects were observed for female rats (1h: t = 0.04, df = 10,
p = 0.968; 1day: t = 0.65, df = 7, p = 0.534; 5days: t = 0.24,
df = 12, p = 0.812; Fig.4D).
Fig. 3 Evaluating the effects
of desipramine on the rate of
hippocampal cell prolifera-
tion in male and female rats as
measured 1-h and 1-day
post-treatment by immunohis-
tochemistry. A Quantitative
analysis of Ki-67 + cells in the
left dentate gyrus, performed
in a light microscope with a
63 × lens. Groups of treatment
(i.p., 3 pulses within a 24-h
period): control (C) (0.9% NaCl,
1ml/kg: 1h, n = 12 males and 6
females; 1day, n = 11 males and
4 females), desipramine (DMI)
10mg/kg (1h: n = 6 males;
1day: n = 6 males) or 20mg/
kg (1h: n = 6 males and 6
females; 1day: n = 6 males and
5 females). Columns represent
mean ± SEM of Ki-67 + cells
(individual symbols are shown
for each rat). One-way ANO-
VAs did not detect any signifi-
cant changes. B Representative
images showing individual
Ki-67 + cells (brown labeling in
the blue granular layer) taken
with a light microscope using
a 40 × objective lens to identify
individual cells
632 S.Ledesma-Corvi, M.J.García-Fuster
1 3
Discussion
This study compared the course of the antidepressant-like
potential of different doses of desipramine administration
and deepened in the putative sex differences in neuroplasti-
city signaling partners modulated in the hippocampus, a key
region for affect regulation. The main results showed that
desipramine induced antidepressant-like effects of similar
magnitude and duration for both sexes (effective doses of
10 and 20mg/kg, with responses that lasted up to 1-day
post-treatment), without altering the rate of cell prolifera-
tion. However, desipramine regulated some neuroplasticity
markers exclusively in male rats (FADD, Cdk5 and p25), in
a way that suggested neuroprotective actions, proposing that
distinctive molecular mechanisms might be participating in
the therapeutic response of desipramine for both sexes.
Fig. 4 Evaluating the effects of
desipramine on hippocampal
neuroplasticity markers in male
and female rats as measured 1-h
and 1- and 5-day post-treatment
by western blot. A FADD, B
Cdk5, C p35 and D p25 protein
content in hippocampal sam-
ples. Groups of treatment (i.p.,
3 pulses within a 24-h period):
control (C) (0.9% NaCl, 1ml/
kg; 1h: n = 12 males and 6
females; 1day: n = 11 males and
4 females), desipramine (DMI)
10mg/kg (1h: n = 6 males;
1day: n = 6 males) or 20mg/
kg (1h: n = 6 males and 6
females; 1day: n = 6 males and
5 females). Columns represent
mean ± SEM of n experiments
per group and expressed as %
control (individual values are
shown for each rat in symbols).
One-way ANOVAs at each
timepoint of analysis followed
by Dunnett's multiple com-
parisons tests when appropri-
ate: *p < 0.05, **p < 0.01 and
*p < 0.05 vs. control rats at the
indicated timepoint of analysis.
E Representative immunoblots
depicting labeling of target
proteins at each timepoint of
analysis
633
Revisiting theantidepressant‑like effects ofdesipramine inmale andfemale adult rats: sex…
1 3
The antidepressant-like response of desipramine was
observed with a similar magnitude in male and female
rats, as decreased rates of immobility and increased climb-
ing in the forced-swim test, therefore, presenting preclini-
cal evidence of efficacy for both sexes. The effective doses
were 10 and 20mg/kg (3 pulses within 24h) and efficacy
was observed 1h and up to 1-day post-treatment. Our data
extended the published literature in the field by showing the
time-course of the antidepressant-like response, since most
previous studies only evaluated a single timepoint (mostly
1-h post-treatment) while also including efficacious doses
for female rats. In particular, desipramine induced a potent
antidepressant-like response as early as 1-h post-treatment,
which was still present with a similar magnitude 1day later,
but dissipated 3-day post-treatment. Some prior studies
suggested that lower doses of desipramine were ineffec-
tive when administered acutely (2 doses of 1, 2 or 5mg/kg
over 24-h, test performed 1-h post-injection), but induced
an antidepressant-like effect only when given chronically
(5mg/kg, 15 injections [18]) in male Sprague–Dawley rats.
However, other acute studies with a higher dose range (5,
10 and 20mg/kg × 3 doses in 24h), similar to the one evalu-
ated here, also demonstrated diminished immobility in the
forced-swim test in male rats [56] (see [19] for our own prior
results with 3 × 20mg/kg). As for female rats, only a few
preclinical studies included females; while one demonstrated
a significant antidepressant-like response with a 3 × 10mg/
kg dose in ovariectomized rats [23], the other one showed
no effects for female Sprague–Dawley rats, even though it
tested doses up to 3 × 10mg/kg, which were effective in
the present study [24]. The authors of that study discussed
that their experimental groups had a limited sample size that
might have prevented observing subtle differences (see [24]).
Interestingly, as expected for antidepressants that enhance
norepinephrine neurotransmission [18, 56, 57], desipramine
increased climbing in the forced-swim test for male rats, but
interestingly, the same pattern was also observed for female
rats. Therefore, contrary to other treatments that had clearly
shown sex differences in antidepressant-like responses, with
a drop in efficacy and/or even an inefficacious response for
females (i.e., electroconvulsive therapy, see [11]; or more
novel compounds, such as ketamine and cannabidiol, see
[12, 13]), desipramine has proven a similar behavioral
response in male and female rats.
In terms of the potential molecular mechanisms mediat-
ing the therapeutic response, the study evaluated an early
stage of hippocampal neurogenesis (cell proliferation,
Ki-67 + cells), but showed no changes for either sex. These
negative effects were consistent with prior findings sug-
gesting the need for a repeated drug treatment (14days) to
promote hippocampal cell proliferation [21, 32]. So far, the
results suggested that while acute desipramine is sufficient to
induce an antidepressant-like response of similar magnitude
for males and female rats, longer treatments might be needed
to increase cell proliferation, suggesting this mechanism
could not fully mediate and/or explain the behavioral out-
come. In this line of thought, previous studies with other
antidepressants, both from our group [47] and others [58],
showed a dissociation between emotional behavior and neu-
rogenesis regulation, suggesting that these responses might
occur independently or, at least, at different time-courses
and/or lengths of treatment. These results contradict prior
studies suggesting the need of a neurogenesis activation for
the antidepressant-like response to occur [34], or for the
observed parallel regulation of both effects [5961], indi-
cating that other factors/molecular players might partici-
pate in the observed antidepressant-like response induced
by desipramine.
In this context, we evaluated the regulation of several key
hippocampal cell-fate markers. Overall, the present results
complemented prior data from our group showing that acute
desipramine induced neuroprotective actions by decreasing
FADD 1-h post-treatment in rat brain cortex [19], to include
another brain region (hippocampus) regulated in a similar
fashion, as well as the course of FADD regulation (up to
5-day post-treatment), and the lack of effects observed in
female rats. Following the initial decrease in FADD 1-h
post-treatment, desipramine increased FADD content 1-day
post-treatment (i.e., rebound effect after the acute initiation
of an anti-apoptotic response, see prior similar results by
[62]) and all levels were normalized 5-day post-treatment,
suggesting a role for this neuroplasticity marker in the effects
induced by desipramine. Moreover, this is the first study
evaluating FADD hippocampal regulation by desipramine
in female rats, proving sex disparities in its regulation that
deserve further characterization.
As for the rest of the hippocampal markers evaluated
(Cdk5, p35 and p25), some indications of neuroprotective
actions were also observed as measured 1-h post-treatment
exclusively in male rats, such as the decrease in neuro-
toxic p25 marker. Interestingly, these effects were also
present 1-day post-treatment, together with an increase
in hippocampal Cdk5. These effects normalized 5-day
post-treatment, when the behavioral response dissipated.
In this context, the activation of the apoptotic-pathway
through FasL, in which FADD is the intracellular adaptor
to Fas receptor, increased the transcription of p35 through
ERK1/2 pathway [63], thus suggesting an indirect connec-
tion for FADD and Cdk5, p35–p25 signaling molecules.
In addition, the regional inhibition of Cdk-5 in the den-
tate gyrus blocked depressive-like behavior (induced by
chronic mild stress) in rats, whereas the overexpression
of p35 blocked the antidepressant-like effect induced by
venlafaxine [43], suggesting a key role for the complex
Cdk5/p35–p25 in the regulation of antidepressant-like
responses. Moreover, Cdk-5 activity was not required for
634 S.Ledesma-Corvi, M.J.García-Fuster
1 3
the proliferation or differentiation of neuronal stem cells,
but was essential for the survival, migration and matura-
tion of newborn granule cells [64]. Overall, the molecular
results suggested the involvement of FADD, Cdk5 and
p35–p25 complex in the neuroplastic actions taking place
in the hippocampus [26] and mediated by desipramine but
exclusively in male rats, demonstrating that although the
behavioral effects were similar, clear sex differences were
observed in the molecular events driving those responses,
that deserve further study.
Taken together, these results showed that while acute
desipramine was capable of inducing antidepressant-like
effects of the same magnitude and duration in male and
female rats, the regulation of some neuroplasticity mark-
ers, that paralleled the behavioral response, was only
detected in male rats. Overall, the results suggested that
the regulation of FADD, Cdk-5 and p35–p25 proteins
could be common markers mediating some of the early
mechanisms taking place right after drug exposure and up
to 1-day post-treatment while matching the course of the
observed behavioral response. On the contrary, the regu-
lation of an early stage of adult hippocampal neurogen-
esis, such as cell proliferation, might require a prolonged
treatment exposure to desipramine, since no effects were
observed following this acute paradigm. Overall, these
molecular correlates were exclusively regulated in male
rats, suggesting other markers might be participating in
the behavioral response observed in females. Further stud-
ies are required to complement these results and deepen
into the neurochemical differences driven by sex in the
response mediated by desipramine, since these dispari-
ties might be responsible for the poor clinical translation
occasionally described.
Acknowledgements We would like to thank Drs. Huda Akil and Stan-
ley J. Watson (University of Michigan, Ann Arbor, MI, USA) who
kindly provided Ki-67 antibody.
Author contributions MJG-F was responsible for the study concept
and design with the contribution of SL-C. SL-C conducted the experi-
ments, analyzed the behavioral and molecular data and drafted the fig-
ures. MJG-F revised and interpreted all the data, as well as reformatted
the figures. MJG-F wrote the manuscript. Both authors have critically
contributed and have approved the final version of the manuscript for
publication.
Funding Open Access funding provided thanks to the CRUE-CSIC
agreement with Springer Nature. Funding for this study was provided
by Grants SAF2014-55903-R and PID2020-118582RB-I00 (MCIN/
AEI/10.13039/501100011033), as well aspartially by PDR2020/14
(Comunitat Autònoma de les Illes Balears through the Direcció General
de Política Universitària i Recerca with funds from the Tourist Stay Tax
Law ITS 2017–006), and Fundación Alicia Koplowitz to MJG-F; these
entities had no further role in study design; in the collection, analysis
and interpretation of data; in the writing of the report; and in the deci-
sion to submit the paper for publication. The ‘García-Palmer’ Summer
Research Program (2019) and JUNIOR Program (IdISBa, GOIB) sup-
ported SL-C’s salary.
Declarations
Conflict of interest The authors have nothing to disclose.
Open Access This article is licensed under a Creative Commons Attri-
bution 4.0 International License, which permits use, sharing, adapta-
tion, distribution and reproduction in any medium or format, as long
as you give appropriate credit to the original author(s) and the source,
provide a link to the Creative Commons licence, and indicate if changes
were made. The images or other third party material in this article are
included in the article's Creative Commons licence, unless indicated
otherwise in a credit line to the material. If material is not included in
the article's Creative Commons licence and your intended use is not
permitted by statutory regulation or exceeds the permitted use, you will
need to obtain permission directly from the copyright holder. To view a
copy of this licence, visit http:// creat iveco mmons. org/ licen ses/ by/4. 0/.
References
1. Kessler RC. Epidemiology of women and depression. J Affect
Disord. 2003;74:5–13.
2. Marcus SM, Young EA, Kerber KB, Kornstein S, Farabaugh AH,
Mitchell J, Wisniewski SR, Balasubramani GK, Trivedi MH, Rush
AJ. Gender differences in depression: findings from the STAR*D
study. J Affect Disord. 2005;87:141–50.
3. Eid RS, Gobinath AR, Galea LAM. Sex differences in depression:
Insights from clinical and preclinical studies. Prog Neurobiol.
2019;176:86–102.
4. LeGates TA, Kvarta MD, Thompson SM. Sex differences in anti-
depressant efficacy. Neuropsychopharmacology. 2019;44:140–54.
5. Herzog DP, Wegener G, Lieb K, Müller MB, Treccani G. Decod-
ing the mechanism of action of rapid-acting antidepressant treat-
ment strategies: Does gender matter? Int J Mol Sci. 2019;20:E949.
6. Kokras N, Dalla C, Papadopoulou-Daifoti Z. Sex differences in
pharmacokinetics of antidepressants. Expert Opin Drug Metab
Toxicol. 2011;7:213–26.
7. Bigos KL, Pollock BG, Stankevich BA, Bies RR. Sex differences
in the pharmacokinetics and pharmacodynamics of antidepres-
sants: an updated review. Gend Med. 2009;6:522–43.
8. Docherty JR, Stanford SC, Panattieri RA, Alexander SP, Cirino
G, George CH, Hoyer D, Izzo AA, Ji Y, Lilley E, Sobey CG,
Stanley P, Stefanska B, Stephens G, Teixeira M, Ahluwalia A.
Sex: a change in our guidelines to authors to ensure that this is
no longer an ignored experimental variable. Br J Pharmacol.
2019;176:4081–6.
9. Miller LR, Marks C, Becker JB, Hurn PD, Chen WJ, Woodruff
T, McCarthy MM, Sohrabji F, Schiebinger L, Wetherington CL,
Makris S, Arnold AP, Einstein G, Miller VM, Sandberg K, Maier
S, Cornelison TL, Clayton JA. Considering sex as a biological
variable in preclinical research. FASEB J. 2017;31:29–34.
10. Beltz AM, Beery AK, Becker JB. Analysis of sex differences in
pre-clinical and clinical data sets. Neuropsychopharmacology.
2019;44:2155–8.
11. García-Cabrerizo R, Ledesma-Corvi S, Bis-Humbert C, García-
Fuster MJ. Sex differences in the antidepressant-like potential of
repeated electroconvulsive seizures in adolescent and adult rats:
Regulation of the early stages of hippocampal neurogenesis. Eur
Neuropsychopharmacol. 2020;41:132–45.
635
Revisiting theantidepressant‑like effects ofdesipramine inmale andfemale adult rats: sex…
1 3
12. Ledesma-Corvi S, García-Fuster MJ. Antidepresssant-like effects
of cannabidiol in a rat model of early-life stress: sex- and age-
dependent efficacy. European College of Neuropsychopharma-
cology Workshop. 2022. Abstract number: WS22–0049. Nice,
France, 17–20 March, 2022.
13. Ledesma-Corvi S, Hernández-Hernández E, García-Fuster MJ.
Antidepressant-like effects of ketamine in a rat model of early-life
stress: sex- and age-dependent efficacy. 34th European College of
Neuropsychopharmacology Congress (ECNP) Congress Hybrid.
2021. 2–5 October, 2021. Lisbon, Portugal.
14. Friedman RA, Kocsis JH. Pharmacotherapy for chronic depres-
sion. Psychiatric Clin of North Am. 1996;19:121–32.
15. Cipriani A, Brambilla P, Furukawa TA, Geddes J, Gregis M,
Hotopf M, etal. Fluoxetine versus other types of pharmacotherapy
for depression. Cochrane Database Syst Rev. 2005;4:CD004185.
16. Cohen LG, Biederman J, Wilens TE, etal. Desipramine clearance
in children and adolescents: absence of effect of development and
gender. J Am Acad Child Adolesc Psychiatry. 1999;38:79–85.
17. Filip V, Alda M, David I, Topinka J, Kristofiková Z, Dvorák-
ová J, Sztaniszláv D, Olájos S, Albrecht V. Neuroendocrine
response to clomipramine and desipramine: the evidence of
partial determination by heredity and sex. Neuropsychobiol-
ogy. 1989;21:111–6.
18. Detke MJ, Johnson J, Lucki I. Acute and chronic antidepressant
drug treatment in the rat forced swimming test model of depres-
sion. Exp Clin Psychopharmacol. 1997;5:107–12.
19. García-Fuster MJ, García-Sevilla JA. Effects of anti-depressant
treatments on FADD and p-FADD protein in rat brain cortex:
enhanced anti-apoptotic p-FADD/FADD ratio after chronic
desipramine and fluoxetine administration. Psychopharmacology.
2016;233:2955–71.
20. Olivares-Nazario M, Fernández-Guasti A, Martínez-Mota L. Age-
related changes in the antidepressant-like effect of desipramine
and fluoxetine in the rat forced-swim test. Behav Pharmacol.
2016;27:22–8.
21. Chen H, Pandey GN, Dwivedi Y. Hippocampal cell proliferation
regulation by repeated stress and antidepressants. NeuroReport.
2006;17:863–7.
22. Slattery DA, Cryan JF. Using the rat forced swim test to
assess antidepressant-like activity in rodents. Nat Protoc.
2012;7:1009–14.
23. Shah A, Frazer A. Influence of acute or chronic administration of
ovarian hormones on the effects of desipramine in the forced swim
test in female rats. Psychopharmacology. 2014;231:3685–94.
24. Simpson J, Ryan C, Curley A, Mulcaire J, Kelly JP. Sex differ-
ences in baseline and drug-induced behavioural responses in clas-
sical behavioural tests. Prog Neuropsychopharmacol Biol Psychia-
try. 2012;37:227–36.
25. Liu W, Ge T, Leng Y, Pan Z, Fan J, Yang W, Cui R. The role of
neural plasticity in depression: from hippocampus to prefrontal
cortex. Neural Plast. 2017;2017:6871089.
26. Tartt AN, Mariani MB, Hen R, Mann JJ, Boldrini M. Dysregula-
tion of adult hippocampal neuroplasticity in major depression:
pathogenesis and therapeutic implications. Mol Psychiatry. 2022.
https:// doi. org/ 10. 1038/ s41380- 022- 01520-y.
27. Jacobs BL, Van Praag H, Gage FH. Adult brain neurogenesis
and psychiatry: a novel theory of depression. Mol Psychiatry.
2000;5:262–9.
28. Malberg JE, Eisch AJ, Nestler EJ, Duman RS. Chronic antidepres-
sant treatment increases neurogenesis in adult rat hippocampus. J
Neurosci. 2000;20:9104–10.
29. Duman RS, Nakagawa S, Malberg J. Regulation of adult neuro-
genesis by antidepressant treatment. Neuropsychopharmacology.
2001;25:836–44.
30. Malberg JE. Implications of adult hippocampal neurogenesis in
antidepressant action. J Psychiatry Neurosci. 2004;29:196–205.
31. Kulkarni VA, Jha S, Vaidya VA. Depletion of norepinephrine
decreases the proliferation, but does not influence the survival
and differentiation, of granule cell progenitors in the adult rat
hippocampus. Eur J Neurosci. 2002;16:2008–12.
32. Warner-Schmidt JL, Duman RS. VEGF is an essential mediator
of the neurogenic and behavioral actions of antidepressants. Proc
Natl Acad Sci USA. 2007;104:4647–52.
33. Jhaveri DJ, Mackay EW, Hamlin AS, Marathe SV, Nandam LS,
Vaidya VA, Bartlett PF. Norepinephrine directly activates adult
hippocampal precursors via β3-adrenergic receptors. J Neurosci.
2010;30:2795–806.
34. Santarelli L, Saxe M, Gross C, Surget A, Battaglia F, Dulawa
S, Belzung C. Requirement of hippocampal neurogenesis for the
behavioral effects of antidepressants. Science. 2003;301:805–9.
35. Saxe MD, Battaglia F, Wang JW, Malleret G, David DJ, etal.
Ablation of hippocampal neurogenesis impairs contextual fear
conditioning and synaptic plasticity in the dentate gyrus. Proc
Natl Acad Sci USA. 2006;103:17501–6.
36. Duman RS. Depression: a case of neuronal life and death? Biol
Psychiatry. 2004;56:140–5.
37. Huang YY, Peng CH, Yang YP, Wu CC, Hsu WM, Wang HJ,
Chan KH, Chou YP, Chen SJ, Chang YL. Desipramine activated
Bcl-2 expression and inhibited lipopolysaccharide-induced apop-
tosis in hippocampus-derived adult neural stem cells. J Pharmacol
Sci. 2007;104:61–72.
38. Zhong P, Liu X, Zhang Z, Hu Y, Liu SJ, Lezama-Ruiz M, etal.
Cyclin-dependent kinase 5 in the ventral tegmental area regulates
depression-related behaviors. J Neurosci. 2014;34:6352–66.
39. Hellmich MR, Pant HC, Wada E, Battey JF. Neuronal cdc2-like
kinase: A cdc2-related protein kinase with predominantly neu-
ronal expression. Proc Natl Acad Sci USA. 1992;89:10867–71.
40. Tsai LH, Delalle I, Caviness VS Jr, Chae T, Harlow E. p35 is a
neural-specific regulatory subunit of cyclin-dependent kinase 5.
Nature. 1994;371:419–23.
41. Patrick GN, Zukerberg L, Nikolic M, de La Monte S, Dikkes P,
Tsai LH. Conversion of p35 to p25 deregulates Cdk5 activity and
promotes neurodegeneration. Nature. 1999;402:615–22.
42. Lagace DC, Benavides DR, Kansy JW, Mapelli M, Greengard
P, Bibb JA, Eisch AJ. Cdk5 is essential for adult hippocampal
neurogenesis. Proc Natl Acad Sci USA. 2008;105:18567–71.
43. Zhu WL, Shi HS, Wang SJ, Xu CM, Jiang WG, Wang X, Wu P,
Li QQ, Ding ZB, Lu L. Increased Cdk5/p35 activity in the dentate
gyrus mediates depressive-like behaviour in rats. Int J Neuropsy-
chopharmacol. 2012;15:795–809.
44. McGrath JC, Lilley E. Implementing guidelines on reporting
research using animals (ARRIVE etc.): new requirements for
publication in BJP. Br J Pharmacol. 2015;172:3189–3193.
45. Percie du Sert N, Ahluwalia A, Alam S, Avey MT, Baker M, etal.
Reporting animal research: Explanation and elaboration for the
ARRIVE guidelines 2.0. PLoS Biol. 2020;18:e3000411.
46. Kokras N, Antoniou K, Mikail HG, Kafetzopoulos V, Papadopou-
lou-Daifoti Z, Dalla C. Forced swim test: What about females?
Neuropharmacology. 2015;99:408–21.
47. Bis-Humbert C, García-Cabrerizo R, García-Fuster MJ. Decreased
sensitivity in adolescent versus adult rats to the antidepressant-like
effects of cannabidiol. Psyhopharmacology. 2020;237:1621–31.
48. Bis-Humbert C, García-Cabrerizo R, García-Fuster MJ. Dose-
dependent opposite effects of nortriptyline on affective-like
behavior in adolescent rats: Comparison with adult rats. Eur J
Pharmacol. 2021;910: 174465.
49. Abel EL. Circannual changes in the duration of the immo-
bility response of rats in the forced-swim test. Physiol Behav.
1995;58:591–3.
50. Bogdanova OV, Kanekar S, D’Anci KE, Renshaw PF. Factors
influencing behavior in the forced swim test. Physiol Behav.
2013;118:227–39.
636 S.Ledesma-Corvi, M.J.García-Fuster
1 3
51. García-Fuster MJ, Perez JA, Clinton SM, Watson SJ, Akil H.
Impact of cocaine on adult hippocampal neurogenesis in an ani-
mal model of differential propensity to drug abuse. Eur J Neuro-
sci. 2010;31:79–89.
52. García-Fuster MJ, Flagel SB, Mahmood ST, Mayo LM, Thomp-
son RC, Watson SJ, Akil H. Decreased proliferation of adult hip-
pocampal stem cells during cocaine withdrawal: possible role
of the cell fate regulator FADD. Neuropsychopharmacology.
2011;36:2303–17.
53. García-Cabrerizo R, Keller B, García-Fuster MJ. Hippocampal
cell fate regulation by chronic cocaine during periods of ado-
lescent vulnerability: consequences of cocaine exposure during
adolescence on behavioral despair in adulthood. Neuroscience.
2015;304:302–15.
54. Curtis MJ, Alexander S, Cirino G, Docherty JR, George CH,
Giembycz MA, etal. Experimental design and analysis and their
reporting II: updated and simplified guidance for authors and peer
reviewers. Br J Pharmacol. 2018;175:987–93.
55. Michel MC, Murphy TJ, Motulsky HJ. New author guidelines for
displaying data and reporting data analysis and statistical methods
in experimental biology. J Pharmacol Exp Ther. 2020;372:136–47.
56. Detke MJ, Rickels M, Lucki I. Active behaviors in the rat forced
swimming test differentially produced by serotonergic and noradr-
energic antidepressants. Psychopharmacology. 1995;121:66–72.
57. Rénéric JP, Bouvard M, Stinus L. In the rat forced swimming
test, chronic but not subacute administration of dual 5-HT/NA
antidepressant treatments may produce greater effects than selec-
tive drugs. Behav Brain Res. 2002;136:521–32.
58. Schiavon AP, Bonato JM, Milani H, Guimarães FS, Weffort
de Oliveira RM. Influence of single and repeated cannabidiol
administration on emotional behavior and markers of cell prolif-
eration and neurogenesis in non-stressed mice. Prog Neuropsy-
chopharmacol Biol Psychiatry. 2016;64:27–34.
59. Madsen TM, Treschow A, Bengzon J, Bolwig TG, Lindvall O,
Tingström A. Increased neurogenesis in a model of electrocon-
vulsive therapy. Biol Psychiatry. 2000;47:1043–9.
60. Malberg JE, Duman RS. Cell proliferation in adult hippocampus is
decreased by inescapable stress: reversal by fluoxetine treatment.
Neuropsychopharmacology. 2003;28:1562–71.
61. Warner-Schmidt JL, Duman RS. Hippocampal neurogenesis:
opposing effects of stress and antidepressant treatment. Hip-
pocampus. 2006;16:239–49.
62. García-Fuster MJ, Clinton SM, Watson SM, Watson SJ, Akil H.
Effect of cocaine on Fas-associated protein with death domain
in the rat brain: individual differences in a model of differen-
tial vulnerability to drug abuse. Neuropsychopharmacology.
2009;34:1123–34.
63. Desbarats J, Birge RB, Mimouni-Rongy M, Weinstein DE,
Palerme JS, Newell MK. Fas engagement induces neurite growth
through ERK activation and p35 upregulation. Nat Cell Biol.
2003;5:118–25.
64. Jessberger S, Aigner S, Clemenson GD Jr, Toni N, Lie DC, Kara-
lay O, etal. Cdk5 regulates accurate maturation of newborn gran-
ule cells in the adult hippocampus. PLoS Biol. 2008;6: e272.
Publisher's Note Springer Nature remains neutral with regard to
jurisdictional claims in published maps and institutional affiliations.
... Previous evidence has depicted this signaling pathway, and particularly FADD, as a key molecule that controls cell fate by balancing apoptotic versus non-apoptotic functions (e.g., Park et al., 2005;Tourneur & Chiocchia, 2010), therefore suggesting a key role for this protein in balancing neurotoxic versus neuroplastic events taking place in the brain under pathophysiological conditions. In this context and over the past 15 years our research group has extensively worked on characterizing the role of FADD in post-mortem brain samples of subjects with psychopathologies (e.g., major depression: García-Fuster et al., 2014;drug addiction: García-Fuster et al., 2008) and/or clinical dementia (e.g., Ramos-Miguel et al., 2017), as well as in the brains of animal models capturing certain aspects of these disorders, in physiological processes such as aging (Hernández-Hernández et al., 2018a), and through the administration of several pharmacological agents (e.g., García-Cabrerizo & García-Fuster, 2019;García-Fuster & García-Sevilla, 2015García-Fuster et al., 2007, 2009Hernández-Hernández et al., 2018a, 2018bLedesma-Corvi & García-Fuster, 2022). The main results presented FADD as a great pharmacological target since treatments with prototypical drugs for these disorders reverted, in some cases, FADD brain changes in key brain regions (mainly studied in prefrontal cortex, striatum and hippocampus) (some data reviewed by . ...
... Additionally, cyclin-dependent kinase-5 (Cdk-5) has been ascribed several functions in the nervous system, ranging from neuronal migration, neuronal outgrowth, axonal guidance, and synaptic plasticity (reviewed by Pao & Tsai, 2021), and has been shown to be modulated in parallel to FADD following desipramine treatment (see Ledesma-Corvi & García-Fuster, 2022), and to crosstalk with MEK-ERK signaling (e.g., . Also, Cdk-5, which has been implicated in the regulation of circadian clocks under physiological conditions (e.g., Brenna et al., 2019;Kwak et al., 2013;Ripperger et al., 2022), is known to depend on co-factors p35 and p25 for its activity (Patrick et al., 1999;Tsai et al., 1994). ...
... These effects might be mediated by the phosphorylation of certain proteins involved in regulating the 24-h clock, since Cdk-5 has been shown to regulate the function of CLOCK (Kwak et al., 2013) and/or that of Period 2 (PER2; Brenna et al., 2019) proteins by direct phosphorylation. These data support prior reports suggesting that Cdk-5 is critically involved in the regulation of the circadian clock and may represent a link to various diseases affected by a derailed daily regulation, probably through the activation of its co-factor p25, and in parallel to FADD regulation (e.g., Ledesma-Corvi & García-Fuster, 2022). ...
Article
Fas‐Associated protein with Death Domain (FADD), a key molecule controlling cell fate by balancing apoptotic versus non‐apoptotic functions, is dysregulated in post‐mortem brains of subjects with psychopathologies, in animal models capturing certain aspects of these disorders, and by several pharmacological agents. Since persistent disruptions in normal functioning of daily rhythms are linked with these conditions, oscillations over time of key biomarkers, such as FADD, could play a crucial role in balancing the clinical outcome. Therefore, we characterized the 24‐h regulation of FADD (and linked molecular partners: p‐ERK/t‐ERK ratio, Cdk‐5, p35/p25, cell proliferation) in key brain regions for FADD regulation (prefrontal cortex, striatum, hippocampus). Samples were collected during Zeitgeber time (ZT) 2, ZT5, ZT8, ZT11, ZT14, ZT17, ZT20, and ZT23 (ZT0, lights‐on or inactive period; ZT12, lights‐off or active period). FADD showed similar daily fluctuations in all regions analyzed, with higher values during lights off, and opposite to p‐ERK/t‐ERK ratios regulation. Both Cdk‐5 and p35 remained stable and did not change across ZT. However, p25 increased during lights off, but exclusively in striatum. Finally, no 24‐h modulation was observed for hippocampal cell proliferation, although higher values were present during lights off. These results demonstrated a clear daily modulation of FADD in several key brain regions, with a more prominent regulation during the active time of rats, and suggested a key role for FADD, and molecular partners, in the normal physiological functioning of the brain's daily rhythmicity, which if disrupted might participate in the development of certain pathologies.
... Our protocol modified the standard protocol [23] in a way that would allow us to evaluate the progression of the response over time. We have done this procedure reliably in the past years as detailed in various publications [9,[24][25][26][27][28][29][30][31][32][33][34], in which to account for the potential emerging effects due to repetition on learning and/or tolerance to test performance, all rats were exposed to the same conditions. In particular, each rat was placed in individual tanks (41 cm high × 32 cm diameter, 25 cm depth; specific dimensions that ensure that the animal does not touch the bottom of the cylinder with its tail) filled with water (25 ± 1 °C) during 15 min (pre-test sessions) followed by 5-min test sessions that were videotaped (see Fig. 1). ...
Article
Full-text available
Background: Ketamine has been recently approved to treat resistant depression; however preclinical studies showed sex differences in its efficacy. Sex steroids, such as estrogens and testosterone, both in the periphery and locally in the brain, are regarded as important modulators of these sex differences. Therefore, the present study evaluated how inhibiting the biosynthesis of estrogens with letrozole (an aromatase inhibitor) could affect the observed sex differences in ketamine's antidepressant-like-response. Methods: We performed several consecutive studies in adult Sprague-Dawley rats to evaluate potential sex differences in the antidepressant-like effects of ketamine (5 mg/kg, 7 days, i.p.), letrozole (1 mg/kg, 8 days, i.p.) and their combination (letrozole pre-treatment 3 h before ketamine). Acute and repeated antidepressant-like responses were ascertained in a series of behavioral tests (forced-swim, novelty-suppressed feeding, two-bottle choice for sucrose preference). Results: The main results proved clear sex differences in the antidepressant-like response induced by ketamine, which was observed following a repeated paradigm in adult male rats, but rendered inefficacious in female rats. Moreover, decreasing estrogens production with letrozole induced on itself an antidepressant-like response in female rats, while also increased ketamine's response in male rats (i.e., quicker response observed after only a single dose). Interestingly, both the antidepressant-like effects induced by ketamine in male rats or letrozole in female rats persisted over time up to 65 days post-treatment, suggesting long-term sex-directed benefits for these drugs. Conclusions: The present results demonstrated a sex-specific role for aromatase inhibition with letrozole in the antidepressant-like response induced by ketamine in male rats. Moreover, letrozole itself presented as a potential antidepressant for females with persistent effects over time. Clearly, the production of estrogens is key in modulating, in a sex-specific manner, affective-like responses and thus deserve further studies.
... To evaluate the potential antidepressant-like effects of ketamine, letrozole and/or their combination, we first screened the animals under the stress conditions of the forced-swim test [23]. To do so, we followed prior well-established procedures in our group (e.g., [24][25][26]), in which rats were placed in individual tanks (41 cm high x 32 cm diameter, 25 cm depth) filled with water (25 ± 1 °C) during 15 min (pre-test sessions) followed by 5-min test sessions that were videotaped (see Fig. 1). Test sessions were repeated across time in an attempt to evaluate the progression of the response: acute effects (e.g., 30 min post-injection), repeated effects (e.g., 1 h or 1-day post-treatment) and/or long-term effects (e.g., 65 days posttreatment). ...
Preprint
Ketamine has been recently approved to treat resistant depression; however preclinical studies showed sex differences in its efficacy. Sex steroids, such as estrogen and testosterone, both in the periphery and locally in the brain, are regarded as important modulators of these sex differences. Therefore, the present study evaluated the role of inhibiting estrogen biosynthesis with letrozole, an aromatase inhibitor that catalyzes the conversion of androgen into estrogen, in the differential antidepressant-like-response induced by ketamine with sex. We performed several consecutive studies in adult Sprague-Dawley rats to evaluate potential sex differences in the antidepressant-like effects of ketamine (5 mg/kg, 7 days, i.p.), letrozole (1 mg/kg, 8 days, i.p.) and their combination (letrozole pre-treatment 3 h before ketamine). Acute and repeated antidepressant-like responses were ascertained in a series of behavioral tests (forced-swim, novelty-suppressed feeding, two-bottle choice for sucrose preference). The main results proved clear sex differences in the antidepressant-like response induced by ketamine, which was observed following a repeated paradigm in adult male rats, but rendered inefficacious in female rats. Moreover, decreasing estrogen production with letrozole induced on itself an antidepressant-like response in female rats, while also improved ketamine’s response in male rats (i.e., quicker response, only after a single dose). Interestingly, both the antidepressant-like effects induced by ketamine in male rats or letrozole in female rats persisted over time up to 65 days post-treatment, suggesting long-term sex-directed benefits for these drugs. The present results demonstrated a sex-specific role for inhibiting estrogen biosynthesis in the antidepressant-like response induced by ketamine in male rats. Moreover, letrozole presented itself as a potential antidepressant for females with persistent effects over time. Clearly, estrogen production is key in modulating, in a sex-specific manner, affective-like responses and thus deserve further studies.
... We also studied another marker of the apoptotic pathway (Cytochrome c, Cty c) whose expression was altered by drugs of abuse (i.e., cocaine, MDMA) in hippocampus in conjunction with FADD [28,29]. Also, Cyclin-dependent kinase-5 (Cdk5) was evaluated since is key in the regulation of neurogenesis [30] and was shown to be modulated in parallel to FADD in hippocampus (see [31]). Finally, the potential structural damage of ethanol was evaluated at the level of neurofilament proteins (e.g., NF-L) as certain drugs of abuse induced neurotoxicity in male rats by decreasing its hippocampal content (e.g., [29]); it was found hyperphosphorylated in hippocampus in response to ethanol toxicity [32], and its circulating levels were altered in heavy drinking in association with lower gray matter thickness [33]. ...
Article
Full-text available
Background Binge alcohol drinking is considered a prominent risk factor for the development of alcohol-use disorders, and could be model in rodents through the standard two-bottle preference choice test. The goal was to recreate an intermittent use of alcohol during 3 consecutive days each week to ascertain its potential impact on hippocampal neurotoxicity (neurogenesis and other neuroplasticity markers), and including sex as a biological variable, given the well-known sex differences in alcohol consumption. Methods Ethanol access was granted to adult Sprague–Dawley rats for 3 consecutive days per week, followed by 4 days of withdrawal, during 6 weeks, mimicking the most common pattern of intake in people, drinking over the weekends in an intensive manner. Hippocampal samples were collected to evaluate signs of neurotoxicity. Results Female rats consumed significantly more ethanol than males, although intake did not escalate over time. Ethanol preference levels remained below 40% over time and did not differ between sexes. Moderate signs of ethanol neurotoxicity were observed in hippocampus at the level of decreased neuronal progenitors (NeuroD + cells), and these effects were independent of sex. No other signs of neurotoxicity were induced by ethanol voluntary consumption when measured through several key cell fate markers (i.e., FADD, Cyt c, Cdk5, NF-L) by western blot analysis. Conclusions Overall, the present results suggest that even though we modeled a situation where no escalation in ethanol intake occurred across time, mild signs of neurotoxicity emerged, suggesting that even the use of ethanol during adulthood in a recreational way could lead to certain brain harm.
Preprint
Full-text available
While ketamine is already approved for treatment resistant depression in adult patients, its efficacy and safety profile for its use in adolescence still needs further investigations. Preclinical studies proved dose- and sex-dependent effects induced by ketamine during adolescence, but few studies have evaluated the short- and long-term safety profile of ketamine at the doses necessary to induce its antidepressant-like effects. The present study aimed at evaluating the antidepressant-like effects of ketamine (1, 5 or 10 mg/kg; vs. vehicle; 1 vs. 7 days) during adolescence in naive or early-life stressed (i.e., maternal deprivation) rats of both sexes in the forced-swim or novelty-suppressed feeding tests. Safety was evaluated by measuring the psychomotor- and reinforcing-like responses induced by adolescent ketamine. In addition, long-term safety was evaluated in adulthood at the level of cognitive performance, or addiction liability (induced by a challenge dose of ketamine in rats treated with adolescent ketamine). The main results reinforced the potential for ketamine as an antidepressant for adolescence, but at different dose ranges for each sex. However, some safety concerns emerged for adolescent female rats (i.e., signs of sensitization at the dose used as antidepressant) and adult male rats (i.e., addiction liability when re-exposed to ketamine in adulthood), suggesting the need for caution and further research before moving forward the use of ketamine as an antidepressant for adolescence.
Article
Full-text available
Background: We recently showed sex differences in the antidepressant-like potential of electroconvulsive seizures (ECS) in adolescent rats; while it worked for male rats it rendered inefficacious in females. Since sex steroids might be important modulators of these sex disparities, we evaluated the role of estrogens in the differential response induced by adolescent ECS. Moreover, given the literature suggesting certain cognitive sequelae from ECS exposure, we aimed at evaluating its long-term safety profile in adulthood. Methods: Adolescent Sprague-Dawley rats were pretreated with letrozole (1 mg/kg/day) or vehicle (1 ml/kg/day) for 8 days (i.p.), and treated during the last 5 days (3-h later) with ECS (95 mA, 0.6 s, 100 Hz) or SHAM. Antidepressant-like responses were measured in the forced swim-test, and long-term cognitive performance was assessed in the Barnes maze. Results: During adolescence, while ECS only exerted an antidepressant-like response in male rats, its combination with letrozole permitted ECS to also induce efficacy in females. Moreover, adolescent ECS treatment improved cognitive performance in adulthood, although exclusively in male rats. Conclusions: Adolescent ECS demonstrated an antidepressant-like potential together with certain long-term beneficial cognitive effects but exclusively in male rats. For females, efficacy was restricted to a situation in which the biosynthesis of estrogens was reduced. Therefore, estrogens and/or testosterone levels play a crucial role in the sex-disparities induced by ECS in Sprague-Dawley rats. Based on this study, and on the literature supporting its safety, ECS should be encouraged to use in cases of treatment-resistant depression during adolescence, while adhering to sex-specific considerations.
Article
Full-text available
In searching for novel targets to design antidepressants, among the characterized imidazoline receptors (IR), I2 receptors are an innovative therapeutical approach since they are dysregulated in major depressive disorder and by classical antidepressant treatments. In fact, several I2 agonists have been characterized for their antidepressant-like potential, but the results in terms of efficacy are mixed, and were exclusively reported in male rodents. Since there are well-known sex differences in antidepressant-like efficacy, this study characterized the potential effects induced by two I2 drugs, CR4056 (i.e., most promising drug already in phase II clinical trial for its analgesic properties) and B06 (a compound from a new family of bicyclic α-iminophosphonates) under the stress of the forced-swim test in male and female rats exposed to early-life stress. Moreover, some hippocampal neuroplasticity markers related to the potential effects observed were also evaluated (i.e., FADD, p-ERK/ERK, mBDNF, cell proliferation: Ki-67 + cells). The main results replicated the only prior study reporting the efficacy of CR4056 in male rats, while providing new data on its efficacy in females, which was clearly dependent on prior early-life stress exposure. Moreover, B06 showed no antidepressant-like effects in male or female rats. Finally, CR4056 increased FADD content and decreased cell proliferation in hippocampus, without affecting p-ERK/t-ERK ratio and/or mBDNF content. Interestingly, these effects were exclusively observed in female rats, and independently of early-life conditions, suggesting some distinctive molecular underpinnings participating in the therapeutic response of CR4056 for both sexes. In conjunction, these results present CR4056 with an antidepressant-like potential, especially in female rats exposed to stress early in life, together with some neuronal correlates described in the context of these behavioral changes in females.
Article
Full-text available
Age and sex are critical factors for the diagnosis and treatment of major depression, since there is a well-known age-by-sex difference in the prevalence of major depression (being females the most vulnerable ones) and in antidepressant efficacy (being adolescence a less responsive period than adulthood). Although the induction of electroconvulsive seizures (ECS) is a very old technique in humans, there is not much evidence reporting sex- and age-specific aspects of this treatment. The present study evaluated the antidepressant- and neurogenic-like potential of repeated ECS across time in adolescent and adult rats (naïve or in a model of early life stress capable of mimicking a pro-depressive phenotype), while including a sex perspective. The main results demonstrated age- and sex-specific differences in the antidepressant-like potential of repeated ECS, since it worked when administered during adolescence or adulthood in male rats (although with a shorter length in adolescence), while in females rendered deleterious during adolescence and ineffective in adulthood. Yet, repeated ECS increased cell proliferation and vastly boosted young neuronal survival in a time-dependent manner for both sexes and independently of age. Moreover, pharmacological inhibition of basal cell proliferation prevented the antidepressant-like effect induced by repeated ECS in male rats, but only partially blocked the very robust increase in the initial cell markers of hippocampal neurogenesis. Overall, the present results suggest that the induction of the early phases of neurogenesis by ECS, besides having a role in mediating its antidepressant-like effect, might participate in some other neuroplastic actions, opening the path for future studies.
Article
Full-text available
Improving the reproducibility of biomedical research is a major challenge. Transparent and accurate reporting is vital to this process; it allows readers to assess the reliability of the findings and repeat or build upon the work of other researchers. The ARRIVE guidelines (Animal Research: Reporting In Vivo Experiments) were developed in 2010 to help authors and journals identify the minimum information necessary to report in publications describing in vivo experiments. Despite widespread endorsement by the scientific community, the impact of ARRIVE on the transparency of reporting in animal research publications has been limited. We have revised the ARRIVE guidelines to update them and facilitate their use in practice. The revised guidelines are published alongside this paper. This explanation and elaboration document was developed as part of the revision. It provides further information about each of the 21 items in ARRIVE 2.0, including the rationale and supporting evidence for their inclusion in the guidelines, elaboration of details to report, and examples of good reporting from the published literature. This document also covers advice and best practice in the design and conduct of animal studies to support researchers in improving standards from the start of the experimental design process through to publication.
Article
Full-text available
RationaleCannabidiol is a non-psychoactive phytocannabinoid with great therapeutic potential in diverse psychiatric disorders; however, its antidepressant potential has been mainly ascertained in adult rats.Objectives To compare the antidepressant-like response induced by cannabidiol in adolescent and adult rats and the possible parallel modulation of hippocampal neurogenesis.Methods Male Sprague-Dawley rats were repeatedly treated with cannabidiol (3, 10, and 30 mg/kg) or vehicle (1 mL/kg) during adolescence (postnatal days, PND 27-33) or adulthood (PND 141-147) and exposed to 3 consecutive tests (forced swim, open field, two-bottle choice) that quantified behavioral despair, anxiety, and sucrose intake respectively.ResultsCannabidiol induced differential effects depending on the age and dose administered, with a decreased sensitivity observed in adolescent rats: (1) cannabidiol (30 mg/kg) decreased body weight only in adult rats; (2) cannabidiol ameliorated behavioral despair in adolescent and adult rats, but with a different dose sensitivity (10 vs. 30 mg/kg), and with a different extent (2 vs. 21 days post-treatment); (3) cannabidiol did not modulate anxiety-like behavior at any dose tested in adolescent or adult rats; and (4) cannabidiol increased sucrose intake in adult rats.Conclusions Our findings support the notion that cannabidiol exerts antidepressant- and anorexigenic-like effects in adult rats and demonstrate a decreased potential when administered in adolescent rats. Moreover, since cannabidiol did not modulate hippocampal neurogenesis (cell proliferation and early neuronal survival) in adolescent or adult rats, the results revealed potential antidepressant-like effects induced by cannabidiol without the need of regulating hippocampal neurogenesis.
Article
Full-text available
The American Society for Pharmacology and Experimental Therapeutics has revised the Instructions to Authors for Drug Metabolism and Disposition, Journal of Pharmacology and Experimental Therapeutics, and Molecular Pharmacology These revisions relate to data analysis (including statistical analysis) and reporting but do not tell investigators how to design and perform their experiments. Their overall focus is on greater granularity in the description of what has been done and found. Key recommendations include the need to differentiate between preplanned, hypothesis-testing, and exploratory experiments or studies; explanations of whether key elements of study design, such as sample size and choice of specific statistical tests, had been specified before any data were obtained or adapted thereafter; and explanation of whether any outliers (data points or entire experiments) were eliminated and when the rules for doing so had been defined. Variability should be described by S.D. or interquartile range, and precision should be described by confidence intervals; S.E. should not be used. P values should be used sparingly; in most cases, reporting differences or ratios (effect sizes) with their confidence intervals will be preferred. Depiction of data in figures should provide as much granularity as possible, e.g., by replacing bar graphs with scatter plots wherever feasible and violin or box-and-whisker plots when not. This editorial explains the revisions and the underlying scientific rationale. We believe that these revised guidelines will lead to a less biased and more transparent reporting of research findings.
Article
Major depressive disorder (MDD) was previously hypothesized to be a disease of monoamine deficiency in which low levels of monoamines in the synaptic cleft were believed to underlie depressive symptoms. More recently, however, there has been a paradigm shift toward a neuroplasticity hypothesis of depression in which downstream effects of antidepressants, such as increased neurogenesis, contribute to improvements in cognition and mood. This review takes a top-down approach to assess how changes in behavior and hippocampal-dependent circuits may be attributed to abnormalities at the molecular, structural, and synaptic level. We conclude with a discussion of how antidepressant treatments share a common effect in modulating neuroplasticity and consider outstanding questions and future perspectives.
Article
Antidepressant drugs elicit different behavioral and neurochemical responses with age. In fact, the use of antidepressants during adolescence is associated with an increased risk of suicidal thinking, being the best pharmacological treatment during this critical period a matter of constant debate in terms of its risk-benefit outcome. In this regard, the present study compared the effects of nortriptyline (3–10 mg/kg, 7 days) on regulating different aspects of affective-like behavior by screening adolescent and adult Sprague-Dawley rats through several consecutive tests (forced-swim, open field, sucrose preference). Brains were later collected to evaluate hippocampal neurogenesis and mBDNF protein content as potential molecular correlates of the observed behavioral responses. The main results in adolescent rats showed that nortriptyline induced dose-dependent opposite effects: while 3 mg/kg decreased immobility and increased mBDNF (indicative of an antidepressant-like response), 10 mg/kg decreased exploratory time in the open field and mBDNF (suggestive of an anxiogenic-like response). These effects were not associated with changes in neurogenesis regulation. In adult rats, nortriptyline failed to modulate affective-like behavior or the neuroplasticity markers evaluated at the doses tested. In conclusion, clear behavioral and neurochemical differences were observed between adolescent and adult rats in response to nortriptyline treatment. Interestingly, while nortriptyline displayed an antidepressant-like potential at the lowest dose examined in adolescence, a higher dose shifted these results towards a negative outcome, thus reinforcing the need to extreme caution when considering this treatment for our younger population.
Article
The American Society for Pharmacology and Experimental Therapeutics has revised the Instructions to Authors for Drug Metabolism and Disposition, Journal of Pharmacology and Experimental Therapeutics, and Molecular Pharmacology These revisions relate to data analysis (including statistical analysis) and reporting but do not tell investigators how to design and perform their experiments. Their overall focus is on greater granularity in the description of what has been done and found. Key recommendations include the need to differentiate between preplanned, hypothesis-testing, and exploratory experiments or studies; explanations of whether key elements of study design, such as sample size and choice of specific statistical tests, had been specified before any data were obtained or adapted thereafter; and explanations of whether any outliers (data points or entire experiments) were eliminated and when the rules for doing so had been defined. Variability should be described by S.D. or interquartile range, and precision should be described by confidence intervals; S.E. should not be used. P values should be used sparingly; in most cases, reporting differences or ratios (effect sizes) with their confidence intervals will be preferred. Depiction of data in figures should provide as much granularity as possible, e.g., by replacing bar graphs with scatter plots wherever feasible and violin or box-and-whisker plots when not. This editorial explains the revisions and the underlying scientific rationale. We believe that these revised guidelines will lead to a less biased and more transparent reporting of research findings.