ArticlePDF Available

Broad-Spectrum Cannabis Oil Alleviates Behavioral Symptoms Associated with Stress-Related Anxiety and Depression in Mice

Authors:

Abstract and Figures

Background: Posttraumatic stress disorder (PTSD) is a psychiatric condition that manifests through a broad range of symptoms and shares several phenotypes with anxiety and depression. Refractory PTSD affects 10 – 30% of the patients and highlights the need for alternative pharmacotherapy. The suggested involvement of the endocannabinoid system (ECS) with the emotional processes has enlightened the use of Cannabis sp. Then, this study aimed to evaluate the therapeutic effects of a broad-spectrum Cannabis oil on anxiety- and depressive-like behaviors triggered by stressors from combined nature. In addition, this study investigated the effect of the oil on central cannabinoid receptor 1 and serum levels of cytokines, chemokines, and growth factors. Methods: Mice were randomized into five groups (vehicle; Cannabis oil; fluoxetine; single oral dose) and submitted to acute restraint and chronic unpredictable stress. Then, they were behaviorally assessed in the elevated plus-maze test (EPMT), forced swimming test (FST), splash test (ST), and open field test (OFT). The tetrad cannabinoid assay evaluated the central effect of the oil. Serum biomarkers levels were measured by a multiplex bead-based assay. Results: Cannabis oil (0.1 mg/kg, p.o.) significantly reduced the anxiety-like behavior in EPMT in the acute restraint stress model (p < 0.05) as compared to vehicle. Moreover, compared to the vehicle, Cannabis oil significantly reverted the despair and anhedonic-like behaviors in FST (p < 0.05) and ST (p < 0.05), respectively, in chronically stressed mice. Yet, compared to vehicle, therapy with Cannabis oil did not induce cannabinoid-tetrad (p < 0.0001); downregulated granulocyte-macrophage colony-stimulating factor (GM-CSF; p < 0.01) and advanced glycation end-products (RAGE; p < 0.0001); and upregulated vascular endothelial growth factor (VEGF; p < 0.01) serum levels. Conclusion: Altogether, our data suggest the potential of the broad-spectrum Cannabis oil to improve symptoms related to anxiety and depression caused by traumatic events.
Content may be subject to copyright.
Pharmaceutical Sciences, 2022, 28(2), 239-250
doi:10.34172/PS.2021.59
https://ps.tbzmed.ac.ir/
Research Article
Broad-Spectrum Cannabis Oil Alleviates Behavioral Symptoms
Associated with Stress-Related Anxiety and Depression in Mice
*Corresponding Author: Rafael Cypriano Dutra, E-mail: rafaelcdutra@gmail.com & Nádia Rezende Barbosa Raposo, E-mail: nadia.barbosa@uf.br
©2021 e Author(s). is is an open access article and applies the Creative Commons Attribution Non-Commercial License (http://creativecommons.
org/licenses/by-nc/4.0/). Non-commercial uses of the work are permitted, provided the original work is properly cited.
Pollyana Mendonça de Assis1, Eduarda Gomes Ferrarini2,3, Gabriela Mantovani Baldasso2, Rodrigo Sebben Paes2,
Murilo Chaves Gouvêa4, Carlos Espínola Neto Segundo4, Francesca Borrelli5, Marcos Antônio Fernandes Brandão1,
Raaele Capasso6,7, Rafael Cypriano Dutra2,3* , Nádia Rezende Barbosa Raposo1*
1Center of Research and Innovation in Health Sciences (NUPICS), School of Pharmacy, Universidade Federal de Juiz de Fora, Juiz de Fora 36036-330, Brazil.
2Laboratory of Autoimmunity and Immunopharmacology (LAIF), Department of Health Sciences, Campus Araranguá, Universidade Federal de Santa
Catarina, Araranguá 88906-072, Brazil.
3Post-Graduate Program of Neuroscience, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis 88040-900, Brazil.
4Associação Brasileira de Apoio Cannabis e Esperança – ABRACE, Parque Sólon de Lucena, 697, João Pessoa 58028-470, Brazil.
5Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy.
6Department of Agricultural Sciences, University of Naples Federico II, 80055 Portici, Italy.
7Endocannabinoid Research Group, Naples, Italy.
Abstract
Background: Posttraumatic stress disorder (PTSD) is a psychiatric condition that manifests
through a broad range of symptoms and shares several phenotypes with anxiety and depression.
Refractory PTSD aects 10–30% of the patients and highlights the need for alternative
pharmacotherapy. e suggested involvement of the endocannabinoid system (ECS) with the
emotional processes has enlightened the use of Cannabis sp. en, this study aimed to evaluate
the therapeutic eects of a broad-spectrum Cannabis oil on anxiety- and depressive-like
behaviors triggered by stressors from combined nature. In addition, this study investigated the
eect of the oil on central cannabinoid receptor 1 and serum levels of cytokines, chemokines,
and growth factors.
Methods: Mice were randomized into ve groups (vehicle; Cannabis oil; uoxetine; single
oral dose) and submitted to acute restraint and chronic unpredictable stress. en, they were
behaviorally assessed in the elevated plus-maze test (EPMT), forced swimming test (FST),
splash test (ST), and open eld test (OFT). e tetrad cannabinoid assay evaluated the central
eect of the oil. Serum biomarkers levels were measured by a multiplex bead-based assay.
Results: Cannabis oil (0.1 mg/kg, p.o.) signicantly reduced the anxiety-like behavior in EPMT
in the acute restraint stress model (p < 0.05) as compared to vehicle. Moreover, compared to the
vehicle, Cannabis oil signicantly reverted the despair and anhedonic-like behaviors in FST (p
< 0.05) and ST (p < 0.05), respectively, in chronically stressed mice. Yet, compared to vehicle,
therapy with Cannabis oil did not induce cannabinoid-tetrad (p < 0.0001); downregulated
granulocyte-macrophage colony-stimulating factor (GM-CSF; p < 0.01) and advanced glycation
end-products (RAGE; p < 0.0001); and upregulated vascular endothelial growth factor (VEGF;
p < 0.01) serum levels.
Conclusion: Altogether, our data suggest the potential of the broad-spectrum Cannabis oil to
improve symptoms related to anxiety and depression caused by traumatic events.
Article Info
Article History:
Received: 11 June 2021
Accepted: 2 September 2021
ePublished: 10 September 2021
Keywords:
-Anxiety
-Broad-spectrum Cannabis oil
-Cannabis sp
-Depression
-Posttraumatic stress disorder
Introduction
Globally, the knowledge of the real prevalence of
posttraumatic stress disorder (PTSD) is unclear.1 It has
been estimated that 61% to 80% of individuals are going
to experience some type of traumatic event during their
lifetime.2 From those individuals, approximately 5% to
10% will develop PTSD.3 is psychiatric ailment aects
the self and social functions of individuals by a broad range
of symptoms, including the involvement of cognition,
emotion, and mood.4 In these people, PTSD is frequently
comorbid with depression and anxiety. According to the
National Epidemiologic Survey on Alcohol and Related
Conditions, 59% and 35.2% of those who met the criteria
for PTSD also met the criteria for anxiety and depression,
respectively.5
Although there are signicant proportions of people
Mendonça de Assis, et al.
240 | Pharmaceutical Sciences, 2022, 28(2), 239-250
suering from PTSD and its comorbidities, such as
depression and anxiety, the molecular mechanisms
underlying the pathophysiology are still poorly
understood. e current knowledge suggests alterations
in neurogenesis, neurohormonal, and neurotransmitter
functioning.6 Another aspect that has attracted attention
is the probable involvement of the immune system and
its dysregulation in PTSD7-9 and PTSD-related disorders
– anxiety and depression.10 en, a better understanding
of the molecular basis of PTSD is worth for future
development of diagnosis, prognosis, and therapeutic
strategies.
inking about new pharmacotherapy options is
particularly important if considering the challenges faced
by patients with the available drugs. To date, the US Food
and Drug Administration (FDA) has only approved
two selective serotonin reuptake inhibitors for the
treatment of PTSD: sertraline and paroxetine.11 O-label
pharmacotherapy includes uoxetine and venlafaxine.12
However, 10 – 30% of the patients are still refractory
to the conventional prescriptions,13 highlighting the
demand for alternative strategies that are safe and have
good tolerability. In this scenario, the endocannabinoid
system (ECS) has emerged as a promising pharmaceutical
target for the modulation of the synaptic transmission
involved with cognition, stress responses, and emotional
stability,14 besides its connection with the immune system
and neurogenesis.15 Consequently, the therapeutic role of
Cannabis sp. has attracted more attention.
Among the more than 400 compounds identied in
Cannabis sp. so far, delta-9-tetrahydrocannabinol (Δ9-
THC) and cannabidiol (CBD) are the most studied
phytocannabinoids. e psychomimetic eects of Cannabis
sp. have been attributed mainly to Δ9-THC, which is an
agonist for central cannabinoid receptor type 1 (CB1R).
CBD, instead, does not elicit euphoria.16 Indeed, CBD is
known for its pharmacological properties, which include
analgesic and anti-inammatory actions.17 Additionally,
this phytocannabinoid has proved to have neuroprotective,
anxiolytic, antipsychotic, antiemetic, and antioxidant
properties through a multi-target mechanism.18-21 Despite
all the pieces of evidence suggesting the eectiveness of
Cannabis sp. on psychiatric disorders, the FDA has only
approved this compound for children who suer from
Lennox-Gastaut Syndrome and Dravet syndromes.22,23 It
is worth mentioning that the approved drugs consist of
derivatives of isolated phytocannabinoids, which are way
dierent from Cannabis oil. According to the extraction
techniques, broad-spectrum Cannabis oil can be almost
free of Δ9-THC but contains all the phytochemicals found
in the plant, including terpenes, avonoids, and other
phytocannabinoids, such as CBD.24 is composition is
said to contribute to the synergistic eects of Cannabis
sp. and might be an alternative treatment for complex
psychiatric diseases.
Animal models are widely used to research new
treatments for PTSD. It helps to comprehend the
molecular basis of the disease and, consequently, identify
potential targets for new drugs or even drug repositioning.
Using animals in pre-clinical research is also a strategy for
screening new potential drugs to treat PTSD.25 Recently,
Deslauriers et al.26 have reviewed >600 articles to examine
the ability of current rodent models to probe biological
and behavioral phenotypes of PTSD. e authors have
evaluated several paradigms, including the restraint
stress and the chronic unpredictable stress, for their
ecacy in stimulating PTSD-like constructs (learned fear
and extinction, avoidance, reduced motivation/reward,
arousal, and cognitive decits) in addition to biological
and physiological phenotypes associated with PTSD.
All the reviewed paradigms produced lasting eects on
general depression- and/or anxiety measures.26 at said,
behavioral tests are methodological tools that represent
the best approach to measure anxiety- and depressive-like
phenotypes in a PTSD model.27
us, considering the high prevalence of PTSD; the
patients’ refractory to medical treatment; and the suggested
therapeutic potential of Cannabis sp. on psychiatric illness,
this study aimed to evaluate in mice the eects of a broad-
spectrum Cannabis oil on anxiety- and depressive-like
behaviors triggered by stressors from combined nature.
Further, it was investigated the central eects of the oil
on CB1R, as well as its inuence on the serum levels of
cytokines, chemokines, and growth factors.
Materials and Methods
Broad-spectrum Cannabis oil
e broad-spectrum Cannabis oil was produced and
analyzed by the Brazilian Association ABRACE (Associação
Brasileira de Apoio Cannabis Esperança, Paraíba - Brazil)
that is enrolled with the National Register of Legal
Entities (CNPJ) under the number 23.877.015/0001-38.
e chromatographic analysis reported a CBD:Δ9-THC
proportion of 11:1 and total cannabinoids of 40.2% (Figure
1). Regarding the microbiological assessment, the oil was
under the current quality parameters.
Animals
A total of 120 male Swiss mice (30-50 g, 50–90 days of age)
were provided by the breeding unit of the Universidade
Federal de Santa Catarina (UFSC). e animals (maximum
of 12 mice group-housed in clear-transparent plastic cages
with dust-free sawdust bedding) were housed under
regulated conditions that included a 12-h-light/-dark cycle
(articial light at 07:00 a.m. to 07:00 p.m.), controlled
temperature (22 ± 2 °C), and standard food and water
ad libitum. All of the tests were conducted between 8:00
a.m. and 5:00 p.m., and the animals were acclimatized to
the laboratory settings for at least 1 h before testing. Each
animal was used only once throughout the experiments.
ey were randomly assigned before treatment or
behavioral evaluation. All eorts were made to minimize
their suering and reduce the number of animals required
for the experiments. e experiments herein described were
Cannabis Oil for Mood Disorders
Pharmaceutical Sciences, 2022, 28(2), 239-250 | 241
Figure 1. High-performance liquid chromatography (HPLC) analysis of broad-spectrum Cannabis oil. THC (retention time = 18.2 min);
CBD (retention time = 10.6 min). THC: tetrahydrocannabinol; CBD: cannabidiol.
reported in compliance with the ARRIVE guidelines28-30
and approved by the Animal Ethics Committee of the
UFSC (CEUA-UFSC) under protocol 7176240920. All the
experimental procedures were conducted according to the
guidelines of CONCEA and CEUA/UFSC, based on the
3R’s principles: replacement, reduction, and renement. A
blind operator performed both the nociception assessments
and the statistical analysis.
Experimental design
Broad-spectrum Cannabis oil (0.1, 1, 3 and 10 mg/kg) and
uoxetine (10 mg/kg), a selective inhibitor of serotonin
used as the positive control, were dissolved in medium-
chain triglycerides (MCT) (Vitafor®, Araçoiaba da Serra,
São Paulo, Brazil) and saline, respectively, and administered
by oral gavage (p.o.). Fluoxetine has been used and proved
to be eective on pre-clinical models of PTSD27,31,32 and
clinical studies.33,34
e chronic protocol was also featured with an additional
group treated continuously with Cannabis oil at 0.1 mg/
kg (p.o.; 1x/administration; 5-day treatment). is extra
group aimed to verify if repeated treatment could result
in a long-lasting anxiolytic-/antidepressant-like eect. All
drugs were prepared right before the treatment, and the
volume of the administration was 10 ml/kg, 1 h before
the constraint stress or immediately aer the last stressful
stimulus (14-day protocol). e choice of the doses used
was based on pilot experiments (Supplemental Data) or on
previous data described in the literature.35-37
Acute restraint stress-induced paradigm
Five groups of randomized mice were tested through
this protocol: vehicle, uoxetine (10 mg/kg, p.o., 1x/
administration), or broad-spectrum Cannabis oil (0.1, 3,
and 10 mg/kg, p.o., 1x/administration), as shown in Figure
2A. For the acute restraint stress-induced protocol, mice
were rst maintained inside their home cages, with free
access to water and food, during the drug administration
and the period before the restriction (1 h). en, each animal
was removed from the group-house and introduced into a
fenestrated punctured plastic tube (18 cm × 4 cm) placed
in a horizontal, so the normal orientation of the animal’s
body was kept. e animal remained in that position for 7
h, with all physical movements restrained but without any
pain. No food or water was oered to the animal during
the 7-hour period. Aer release, the animals waited for 40
min before being behaviorally assessed.38-41 e researcher
was blind for the treatments, and the behavioral tests were
manually scored.
Chronic unpredictable stress (CUS) paradigm
Five groups of randomized mice were evaluated: vehicle,
uoxetine (10 mg/kg, p.o., 1x/administration), or broad-
spectrum Cannabis oil (0.1, 1 and 3 mg/kg, p.o., 1x/
administration), as shown in Figure 2B. is protocol
was also featured with an additional group that was
continuously treated with Cannabis oil at 0.1 mg/kg (p.o.) –
administered in the last 5 days of the protocol. e stressful
stimuli were designed to maximize the unpredictable
nature of the stressor. erefore, several stressors varying in
duration and time were randomly applied for 14 days, and
food and water were oered ad libitum. As soon as mice
were exposed to the stressor, they returned to their home
cage and were kept under laboratorial standard conditions.
Mice received the aforementioned treatments on the 14th
and last day of the protocol and were behaviorally assessed
24 h aer the treatment.42,43
Briey, the CUS paradigm consisted of exposure, once
daily, to one of the following aversive stressors: restraint –
mice were placed into a plastic tube (18 cm × 4 cm) sealed
at the extremities and properly perforated to promote
air circulation. is stimulus occurred on the 1st and 7th
Mendonça de Assis, et al.
242 | Pharmaceutical Sciences, 2022, 28(2), 239-250
days of the protocol; forced swimming – mice were into
a cylindrical recipient (10 cm × 25 cm) containing 19 cm
of water on the 6th day of the protocol; cold bath – mice
were placed inside a group-house with 2 cm of water on the
2nd and 9th days of the protocol; wet wood shaving – wood
shavings were wet with 400 ml of water, and the group-
house was tilted at 45° angle. is stimulus occurred on the
3rd and 12th days of the protocol; footshock – test took place
at the passive avoidance apparatus (Insight® – Ribeirão
Preto, São Paulo, Brazil). Mice were placed on the bars and
received paw shocks (0.7 mA; 0.5 s/min) every 30 sec for
3 min on the 8th, 10th, and 14th days of the protocol; tail
compression – on the 4th and 13th days, the tail compression
stimulus was conducted by positioning a clothespin 1 cm
from the base of the animal’s tail. Figure 2B summarizes all
the steps aforementioned.
Behavioral tests
Forced swimming test
e FST assessed antidepressant-like behavior and followed
the method described by Porsolt et al.44 Each one of the
animals was placed into a transparent cylindrical tank (30
cm × 20 cm) with 15 cm of water at a temperature of 22-25
ºC. e test was conducted for 6 min, with a habituation
period of 2 min. e antidepressant-like eects of the
treatments were assessed in the function of the latency to
immobility.45,46 e FST was only executed in the chronic
protocol.
Splash test
e splash test (ST) was used to measure the anhedonia-
like state. It consisted of squirting a sucrose solution (200
µl, 20%) on the dorsal coat of the animals. Because of the
high viscosity of sucrose in this concentration, the animals
initiate the self-cleaning behavior, a typical symptom of
anhedonia used to a depression diagnosis. Aer squirting,
the latency to grooming was recorded for 5 min as an
indicator of self-care and motivational behavior.27,47 e ST
was only executed in the chronic protocol.
Elevated plus-maze test
e elevated plus-maze test (EPMT) assessed anxiety-like
behavior in mice. e apparatus comprises two open arms
(36 cm × 5 cm) and two closed arms (36 cm × 5 cm × 18
cm) that are connected to a central platform (5 cm × 5 cm)
and elevated 50 cm from the ground. For the test, an animal
per time was placed at the center of the platform, facing
toward the closed arm, and allowed to move through the
arms. e time spent in the open arms and the number
of entries made into the open arms were recorded for 5
min. Subsequently, the percentage of time spent in open
arms was calculated from the total spent there divided by
the total time spent in both open and closed arms. e
percentage of entries made into the open arms was given
from the total entries made into these arms divided by the
total entries made in both open and closed arms.48
Open eld test
e open-eld test (OFT) evaluated whether the animals
Figure 2. Experimental design. (A) The acute restraint stress-induced protocol was conducted in four different stages as illustrated above
(I-IV), which included the drug administration 1 h before the restraint, the containment in tubes for 7 h, the ambiance of 40 min, and the
behavioral tests followed by euthanasia. Behavioral tasks were evaluated by the elevated plus-maze and open eld tests. (B) The chronic
unpredictable stress (CUS) protocol, applied for 15 days, was divided into periods of stressful stimuli and behavioral tests/euthanasia. The
stressful stimuli consisted of containment, forced swimming, cold bath, wet wool shavings, shock, and tail compression in alternated days
and hours, always unpredictably. In this protocol, the drug was administered on the last day (14) or daily for the group under continuous
treatment. Behavioral tasks were assessed by the forced swimming (FST), open eld (OFT), elevated plus-maze (EPMT), and splash
(ST) tests.
Cannabis Oil for Mood Disorders
Pharmaceutical Sciences, 2022, 28(2), 239-250 | 243
had any locomotor impairment during the experimental
protocols and treatments. Each one of the mice was placed
in the center of an acrylic box (30 cm × 30 cm) that possess
nine square areas equally divided (Insight® – Ribeirão
Preto, São Paulo, Brazil). e crossing number (scored by
the number of segments crossed with the four paws) was
used to assess locomotor activity. e test lasted 5 min per
animal, and the apparatus was cleaned with a solution of
ethanol 10% aer each test to avoid clues and smells from
the predecessor animal.49
Cannabinoid-induced tetrad
e classical cannabinoid-induced tetrad is a preclinical
model that evaluates the “safety-pharmacology” of new
cannabinoid-related molecules.50 e main purpose
of the test was to monitor the central eects of broad-
spectrum Cannabis oil on cannabinoids by measuring the
following parameters: spontaneous locomotor activity,
rectal temperature, catalepsy, and antinociception.51 Four
groups were assessed for each parameter aforementioned:
vehicle (MCT); broad-spectrum Cannabis oil (3 or 30
mg/kg, p.o.); or WIN 55,212 (1.5 mg/kg, i.p. – a potent
cannabinoid receptor agonist). Tests were conducted every
1 h, during the period of 6 h following the drug or vehicle
administration.
Spontaneous locomotor activity was evaluated using
the rotarod apparatus (Ugo Basile, Italy). It was xed at a
rotational speed of 4 revolutions per minute (rpm). Before
the experiment, mice had been trained for 60 sec on two
consecutive days. Latencies were determined in the case
the animals had fallen o the apparatus.
Core temperatures were measured using a clinic digital
thermometer (BD Basics, New Jersey, USA) that was
lubricated with intimate gel before inserting it into the
rectum to a constant depth of 3 cm.
To measure catalepsy mice were hung by their frontal
paws from a plastic stem (12 cm of diameter) xed
horizontally at a height of 2-3 cm, allowing them to stay
standing. is test measured the time that the animal spent
moving and touching the bottom of the box. e test cut-
o point was 180 sec.
To evaluate antinociception, the tail-ick apparatus with
a xed temperature of 45 °C. e test had a cut-o point
of 15 sec.
Serum biomarkers
Blood was collected from each animal on the 15th and
last day of CUS protocol, and serum was separated by
centrifugation (5500 × g for two 15-min cycles) and stored
until further analysis. Briey, 50 µl of the sample was
processed with a multiplex bead-based assay (R&D Systems,
Minneapolis, USA) according to the manufacturer’s
instructions. e assay determined the serum levels of
dierent cytokines, chemokines, and growth factors [for
instance, monocyte chemoattractant protein-1 (MCP-
1), eotaxin (EOTX), granulocyte-macrophage colony-
stimulating factor (GM-CSF), interleukin- (IL-) 1β, IL-4,
IL-17A, IL-33, IL-2, IL-6, IL-17E, vascular endothelial
growth factor (VEGF), macrophage inammatory protein
(MIP)-1α, keratinocytes-derived chemokine (KC),
intercellular adhesion molecule (ICAM), and receptor for
advanced glycation end products (RAGE)]. Measurements
and analysis were performed by the Luminex platform
(Luminex® 100/200™ System, Texas, EUA). Each multiplex
immunoassay was performed in quintuplicate, and results
were expressed as pg/ml serum.
Statistical analysis
All data are expressed as the mean ± standard error of
the mean (SEM) of 4 – 10 animals/group. A statistical
comparison of the data was performed by one- or two-way
ANOVA followed by Bonferroni to multiple comparisons
post hoc test. P < 0.05, 0.01, and 0.001 were considered
signicant. Statistical analyses were performing using
GraphPad Prism 8.2.1 soware (GraphPad Soware Inc.,
San Diego, CA, USA).
Results
Eects of single administration doses of broad-spectrum
Cannabis oil on acute restraint stress-induced behaviors
in the EPMT and OFT
e EPMT assessed the anxiolytic eect of the treatment
with a single dose of broad-spectrum Cannabis oil (0.1, 3,
and 10 mg/kg, p.o.) during acute restraint stress-induced
behaviors. As shown in Figure 3A, the oil signicantly
increased the time spent in open arms at 0.1 mg/kg as
compared to vehicle (p < 0.05). Figure 3B shows that the
more entries made into the open arms by the animals from
the groups treated with Cannabis oil were not signicantly
dierent from the vehicle (p > 0.05). Finally, no changes
were observed in the locomotor activity (p > 0.05) in the
OFT (Figure 3C), conrming that the anxiolytic eect of
Cannabis oil occurs independently of any motor changes.
Compared to the positive group, no signicant changes
were observed.
Eects of the treatment with broad-spectrum Cannabis oil
on CUS-induced behaviors
As shown in Figure 4A, the treatment with broad-spectrum
Cannabis oil at 0.1 mg/kg signicantly diminished the
latency to immobility in the FST as compared to the
vehicle group (p < 0.05), similarly to the eect seen in the
uoxetine group (p < 0.05). Moreover, Figure 4B showed
that mice treated with Cannabis oil (0.1, 1, and 3 mg/kg)
started grooming signicantly faster than animals treated
with vehicle (p < 0.05) in the ST. Relevantly, the OFT
(Figure 4C) showed no change in the locomotor behavior
of the animals (p > 0.05). Regarding the EPMT (Figure
4D, E) no signicant dierences were observed in the time
spent in the open arms (p > 0.05), although mice treated
with Cannabis oil (0.1 mg/kg) signicantly demonstrated
more entries into the open arms (p < 0.01). Finally, mice
under CUS protocol were continuously treated with broad-
spectrum Cannabis oil (0.1 mg/kg) for 5 days and then
Mendonça de Assis, et al.
244 | Pharmaceutical Sciences, 2022, 28(2), 239-250
Figure 3. Effect of the treatment with broad-spectrum Cannabis oil (0.1, 3, and 10 mg/kg, p.o.) or uoxetine (10 mg/kg, p.o.) on mice
induced to acute restraint stress and submitted to EPMT (A and B) and OFT (C). Values are expressed as mean ± SEM of 7-10 animals
per column. ***p < 0.001 versus vehicle (one-way ANOVA followed by Bonferroni post-hoc-test). EPMT: elevated plus-maze test; OFT:
open eld test.
behaviorally compared to vehicle and uoxetine groups,
although no signicant changes were observed (data not
shown).
Broad-spectrum Cannabis oil did not induce cannabinoid-
like eects on tetrad assay
e eects of treatment with broad-spectrum Cannabis
oil (3 and 30 mg/kg, p.o.) on locomotion, nociception,
catalepsy, and body temperature are shown in Figure 5. As
expected, WIN 52,212-2 (1.5 mg/kg, i.p.), a potent CB1R
agonist, induced cannabinoid-like eects during tetrad
assay. is agonist reduced signicantly the locomotor
activity (p < 0.0001; Figure 5A), increased the threshold
sensitivity (p < 0.0001, Figure 5B), induced catalepsy (<
0.0001; Figure 5C), and decreased the body temperature (p
< 0.0001; Figure 5D) as compared to the vehicle. Otherwise,
no signicant behavior changes were observed following
the oral administration of Cannabis oil (p > 0.05).
Serum biomarkers
Table 1 shows the cytokines, chemokines, and growth
factors levels following a single dose or continuous
Figure 4. Effects of the treatment with broad-spectrum Cannabis oil (0.1, 1 and 3 mg/kg; p.o.) or uoxetine (10 mg/kg; p.o.) on mice
chronically stressed by unpredictable stressors and submitted to the FST (A), ST (B), OFT (C), and EPMT (D and E). Treatments were
administered 24 h before behavioral assessments. Values are expressed as mean ± SEM of 7-10 animals per column. *p < 0.05 and **p
< 0.01 versus vehicle (one-way ANOVA followed by Bonferroni post-hoc-test). FST: forced swimming test; ST: splash test; OFT: open eld
test; EPMT: elevated plus-maze test.
Cannabis Oil for Mood Disorders
Pharmaceutical Sciences, 2022, 28(2), 239-250 | 245
Figure 5. Evaluation of the treatment with broad-spectrum Cannabis oil on cannabinoid tetrad assay. The tests included locomotor ac-
tivity (A), threshold hyperalgesia (B), catalepsy-like behavior (C), and thermal body measurement (D). WIN 52,212-2 (1.5 mg/kg, i.p.) – a
potent cannabinoid receptor agonist – was used as the positive control. Data are presented as mean ± SEM of 4-5 mice per group. **p
< 0.001 versus naïve (two-way ANOVA followed by Bonferroni post-hoc-test). B: baseline withdrawal threshold refers to the evaluation
performed before treatment.
Table 1. Cytokines, chemokines, and growth factors serum levels following single and continuous treatment with broad-spectrum Cannabis
oil (0.1 mg/kg, p.o.).
Measurement Vehicle Fluoxetine Single Dose Continuous Treatment P-value
MCP-1 281.5 (12.42) 294.8 (14.27) 340.8 (28.86) 294.9 (24.54) 0.2354
EOTX 255.7 (35.97) 249.9 (16.86) 282.1 (11.32) 256.7 (16.64) 0.7447
GM-CSF 4.525 (0.10) 4.289 (0.10) 4.018 (0.09)** 4.304 (0.08) 0.0081
IL-1β 49.76 (6.89) 51.49 (8.62) 45.40 (9.03) 44.98 (7.39) 0.9244
IL-4 ND ND ND ND -
IL-17A 7.769 (1.55) 6.971 (0.95) 10.55 (2.43) 10.37 (3.43) 0.6031
IL-33 54.86 (1.57) 47.57 (2.10) 56.12 (2.88) 52.04 (2.75) 0.0775
VEGF 12.88 (0.37) 12.77 (0.65) 13.66 (0.54) 22.26 (4.06)** 0.0075
MIP-1α 0.2138 (0.04) 0.2583 (0.08) 0.3688 (0.08) 0.3550 (0.03) 0.1943
KC 65.79 (3.41) 54.06 (2.27) 60.59 (1.82) 68.68 (6.25) 0.0590
ICAM 25450 (2138) 24210 (900.7) 20710 (1517) 22360 (1420) 0.1734
IL-2 3.630 (0.13) 3.423 (0.27) 3.718 (0.22) 3.208 (0.15) 0.3012
IL-6 3.370 4.430 (1.12) 5.810 (1.22) 7.030 0.1596
IL-17E 5.296 (5.296) 5.296 (5.296) 8.406 (5.369) 15.84 (8.925) 0.6196
RAGE 36.40 (1.97) 18.13 (2.12)* 36.62 (1.18) 20.19 (2.98)*** <0.0001
Measurements were in ng/ml. Values are reported as mean ± SEM. *p < 0.05, **p < 0.01, and ***p < 0.001 versus vehicle (one-way ANOVA
followed by Bonferroni post-hoc test). ND: not detected.
treatment with broad-spectrum Cannabis oil (0.1 mg/kg,
p.o.) during CUS protocol. Serum levels of GM-CSF were
signicantly reduced (p < 0.01) when compared to vehicle,
aer single-dose administration. Under continuous
treatment and compared to the vehicle group, serum
levels of VEGF were signicantly upregulated (p < 0.01),
and RAGE levels were signicantly downregulated (p <
0.0001). No statistically signicant dierences were found
for the other measurements. e level of IL-4 could not be
detected in the assay.
Discussion
e present results showed that broad-spectrum Cannabis
oil reduced the anxiety-like behavior triggered by the
acute restraint paradigm. e oil also reverted despair and
anhedonic-like behaviors triggered by unpredictable stress.
Mendonça de Assis, et al.
246 | Pharmaceutical Sciences, 2022, 28(2), 239-250
Importantly, cannabinoid-like eects were not observed at
the doses of 3 and 30 mg/kg of Cannabis oil during tetrad
assay. Lastly, oral administration of the oil downregulated
the serum levels of GM-CSF and RAGE, whereas VEGF
serum levels were found to be upregulated.
Cannabis sativa, a botanical plant with a millenary
history of medicinal use, and its phytocannabinoids have
been under investigation by their potential eects on a
wide range of conditions, including psychiatric illnesses.52
Stressful episodes inuence homeostasis by changing the
physiological and neurobehavioral proles throughout
adaptive processes. Stress is one of the external causes
of anxiety and depression, the two of the most common
psychiatric illnesses, and is known for playing critical roles
in the pathophysiology of these conditions.48,53 Interestingly,
the involvement of the ECS with emotional processing has
gained attention in the last few years, suggesting the use of
Cannabis sp. as a therapeutic alternative for the treatment
of symptoms related to PTSD.52
In the rst set of experiments, mice were treated with
broad-spectrum Cannabis oil and then submitted to the
acute restraint stress protocol. Substantial ndings have
supported this model as able to evoke PTSD-like constructs,
including depressive- and anxiety-like symptoms.26,38,53-56
Moreover, this protocol has been employed to screen the
therapeutic potential of drugs to manage mood symptoms
related to PTSD since behavioral changes in mice can be
monitored. Herein, we found that Cannabis oil (0.1 mg/
kg; p.o.), rich in CBD, showed an anxiolytic eect while
it did not change depressive-like symptoms. Previously,
Resstel et al.57 also demonstrated that a single dose of CBD
(10 mg/kg, i.p.) increased the percentage of open arm
entries in the EPMT of rats that have had their movements
restrained. is anxiolytic action was attributed to the
activation of 5-HT1A receptors. Yet, a recent study showed
that mice exposed to traumatic brain injury had the
anxiety- and depressive-like behaviors reestablished by a
commercially available 10% CBD oil.58 Contradicting these
data and the study of Sales et al.59, which demonstrated the
antidepressant-like eect of CBD (10 mg/kg, i.p.) on mice
in the FST, we found that Cannabis oil was ineective to
revert the depressive-like symptom in the FST. It is worth
mentioning that those studies evaluated depressant-like
symptoms triggered by other protocols as compared to
ours. Yet, we are comparing the results of isolated CBD
with a broad-spectrum oil, even though the pieces of
evidence suggest that there is a positive contribution from
the combination of phytocannabinoid and other molecules,
such as terpenes, also called as “entourage eect”.60
Otherwise, neuropsychiatric alterations related to
anxiety- and depressive-like behaviors are also described
as consequences of exposure to chronic unpredictable
stress.43,61,62 In the second set of experiments, dierent
types of stressors applied to mice for 14 days signicantly
altered the parameters related to latency to immobility
in the FST and grooming behavior in the ST. In rodents,
a reduced persistence of swimming and a sucrose
indierence are commonly associated with depressive-
like symptoms.39 Since we observed an improvement of
the anhedonia and depressive patterns, we could interpret
these data as a consequence of the treatment with broad-
spectrum Cannabis oil. Previously, isolated CBD (10 mg/
kg; i.p.) had exerted pro-hedonic eects on rats subjected
to chronic unpredictable mild stress by increasing their
sucrose preference.39 Still, other reports demonstrated that
CBD isolated (30 and 200 mg/kg) signicantly diminished
depressive-like behavior in mice during FST.63,64 Taken
together, one may conclude that Cannabis and Cannabis
derivatives reduce depressant-like behaviors in rodents in
a large range of dosages.
When evaluating the potential psychoactive eects of
cannabinoids, the tetrad assay is very useful to characterize
their biological activity. Mainly, the cannabinoid tetrad
reveals cannabimimetic eects related to those elicited by
CB1R agonist ∆9-THC.65 Our ndings showed no eects
of broad-spectrum Cannabis oil at 3 and 30 mg/kg in
the tetrad assay, conrming the analytical parameters of
quality attested by the supplier (CBD:Δ9-THC proportion
of 11:1 and total cannabinoids of 40.2%). CBD is well
known for its low anity by CB1R and as expected, it does
not activate the cannabinoid tetrad.66 Our ndings are in
accordance with these previous data since the Cannabis oil
we tested is rich in CBD, but it is not the only compound.
us, the psychoactive eects on mood-related symptoms
appear to be independent of CB1R activation, although
further experiments are needed to conrm this hypothesis.
Earlier experiments have suggested the psychoactive
action of phytocannabinoids, mainly CBD, throughout
the 5-HT1A signaling pathway.57,67 Besides, the pieces of
evidence suggesting the anti-inammatory properties
of Cannabis68,69 contributed to the increasing interest
in its therapeutic potential in mood disorders. Earlier
experiments have demonstrated that GM-CSF levels are
downregulated with phytocannabinoids treatment.70,71
We also found a signicant decrease of GM-CSF serum
levels following a single dose administration of broad-
spectrum Cannabis oil (0.1 mg/kg, p.o.). is chemokine
plays a critical role in regulating leukocyte counts,72 of
which are elevated in PTSD patients.73,74 Another nding
of our study was the signicant downregulation of RAGE
following continuous treatment with broad-spectrum
Cannabis oil (0.1 mg/kg, p.o.). To the light of our current
knowledge, there are no previous reports regarding the
inuence of Cannabis, or any isolated phytocannabinoid,
upon RAGE expression. is receptor is known for its
ability to recognize danger-associated molecular patterns
(DAMPs) that can be released on a larger scale because of
psychological and physical stress.75 A recent review showed
that several DAMPs, including the high mobility group
box-1 (HMGB-1), may trigger depressive-like behaviors in
the stress-induced depression model.76 HMGB-1 is a well-
known ligand of RAGE and, interestingly, a prospective
study had associated the high plasma levels of HMGB-1
with the more likely to develop PTSD.77 Taken together,
Cannabis Oil for Mood Disorders
Pharmaceutical Sciences, 2022, 28(2), 239-250 | 247
the accumulating data suggest the role of the HMGB-
1/RAGE signaling in mood processes, and the eects
observed in our study suggest that Cannabis may mediate
immunomodulatory eects through this cellular pathway.
Last, but not least, the continuous treatment with broad-
spectrum Cannabis oil (0.1 mg/kg, p.o.) signicantly
upregulated the serum levels of VEGF. In dierent cells
and tissues, VEGF plays key roles in physiologic vascular
homeostasis but is also associated with the molecular
pathogenesis of tumor growth and metastasis.78 Previously,
Wheal et al.79 showed the increase of the circulating levels
of VEGF in ZDF diabetic rats treated with CBD, while
Leishman et al.80 showed a signicant lowering of the
mRNA encoding for VEGF in mice following acute Δ9-
THC administration (3 mg/kg; i.p.). We are not aware of
any studies that have investigated the eects of Cannabis
oil, and not only isolated phytocannabinoids, on VEGF
levels in stress-induced mice, thus justifying additional
investigation.
Conclusion
In summary, our data showed the potential of broad-
spectrum Cannabis oil to change behavioral patterns related
to stress-induced anxiety- and depressive-like symptoms.
Importantly, the oil did not trigger psychomimetic eects
related to CB1R activation, as shown by the tetrad assay.
Moreover, our data suggest the potential of the oil to
regulate biomarkers involved with inammation and
angiogenesis, although further investigations are required
to conrm these hypotheses.
Ethical Issues
e experiments were approved by the Animal Ethics
Committee of the UFSC (CEUA-UFSC) under protocol
7176240920.
Acknowledgments
Grants from the Conselho Nacional de Desenvolvimento
Cientíco e Tecnológico (CNPq), Coordenação de
Aperfeiçoamento de Pessoal de Nível Superior (CAPES),
Fundação de Amparo à Pesquisa e Inovação do Estado de
Santa Catarina (FAPESC), Programa INCT-INOVAMED
(Grant no. 465430/2014-7) and Programa de Pós-
Graduação em Neurociências (PPG NEURO), all from
Brazil, supported this work. P.M.A. and E.G.F. are PhD
student on the Health and Neuroscience Program,
respectively, receiving grants from the CAPES. G.M.B. and
R.S.P. are graduate student receiving grant from the CNPq.
R.C.D. is recipient of a research productivity fellowship
from the CNPq. e authors are grateful to the ABRACE
association in Paraíba – Brazil for providing the broad-
spectrum CBD oil obtained from Cannabis sp. used in this
study.
Author Contributions
RCD, NRBR, MAFB, and RC contributed to the
conception and design of the experiments. MCG,
CENS, and FB contributed to the acquisition and
analysis of data. EGF, GMB, and RSP contributed to
the acquisition, analysis, and interpretation of data.
PMA contributed to the analysis and interpretation
of data; writing the manuscript with input from all
authors. RC and NRBR supervised the project and
revised it critically for important intellectual content.
Conict of Interest
e authors report no conicts of interest.
References
1. Ophuis RH, Olij BF, Polinder S, Haagsma JA. Prevalence
of post-traumatic stress disorder, acute stress disorder
and depression following violence related injury treated
at the emergency department: a systematic review.
BMC Psychiatry. 2018;18:311. doi:10.1186/s12888-
018-1890-9
2. Wang Z, Zhu H, Yuan M, Li Y, Qiu C, Ren Z, Yuan C,
Lui S, Gong Q, Zhang W. e resting-state functional
connectivity of amygdala subregions associated with
post-traumatic stress symptom and sleep quality in
trauma survivors. Eur Arch Psychiatry Clin Neurosci.
2021;271(6):1053-64. doi: 10.1007/s00406-020-01104-
3
3. Mann SK, Marwaha R. Posttraumatic Stress Disorder.
[Updated 2021 Jul 7]. In: StatPearls [Internet].
Treasure Island (FL): StatPearls Publishing; 2021 Jan-
.Available from: https://www.ncbi.nlm.nih.gov/books/
NBK559129/
4. Lisieski MJ, Eagle AL, Conti AC, Liberzon I, Perrine
SA. Single-Prolonged Stress: A Review of Two Decades
of Progress in a Rodent Model of Post-traumatic Stress
Disorder. Front Psychiatry. 2018;9:196. doi:10.3389/
fpsyt.2018.0019
5. Orsolini L, Chiappini S, Volpe U, Volpe U, Berardis D,
Latini R, et al. Use of medicinal cannabis and synthetic
cannabinoids in post-traumatic stress disorder (PTSD):
A systematic review. Medicina (Kaunas). 2019;55:525.
doi:10.3390/medicina55090525
6. Aliev G, Beeraka NM, Nikolenko VN, Svistunov AA,
Rozhnova T, Kostyuk S, et al. Neurophysiology and
Psychopathology Underlying PTSD and Recent Insights
into the PTSD erapies-A Comprehensive Review. J
Clin Med. 2020;9(9):2951. doi:10.3390/jcm9092951
7. Wang Z, Caughron B, Young MRI. Posttraumatic
stress disorder: An immunological disorder? Front
Psychiatry. 2017;8:222. doi:10.3389/fpsyt.2017.00222
8. Hori H, Kim Y. Inammation and post-traumatic stress
disorder. Psychiatry Clin Neurosci. 2019; 73(4):143-53.
doi: 10.1111/pcn.12820
9. Kim TD, Lee S, Yoon S. Inammation in post-traumatic
stress disorder (PTSD): A Review of potential correlates
of PTSD with a neurological perspective.Antioxidants
(Basel). 2020;9(2):107. doi:10.3390/antiox9020107
10. Felger JC. Imaging the role of inammation
in mood and anxiety-related disorders. Curr
Mendonça de Assis, et al.
248 | Pharmaceutical Sciences, 2022, 28(2), 239-250
Neuropharmacol. 2018;16(5):533-58. doi:10.2174/157
0159X15666171123201142
11. Sareen J. Posttraumatic stress disorder in adults: impact,
comorbidity, risk factors, and treatment. Can J Psychiat ry.
2014;29:460-7. doi:10.1177/070674371405900902
12. VA/DoD Management of Post-Traumatic Stress
Working Group. VA / DoD Clinical Practice
Guideline For e Management Of Posttraumatic
Stress Disorder and Acute Stress Disorder. https://
www.healthquality.va.gov/guidelines/MH/ptsd/
VADoDPTSDCPGFinal012418.pdf
13. Akiki TJ, Abdallah CG. Are there eective
psychopharmacologic treatments for PTSD? J
Clin Psychiatry. 2018;80:18ac12473. doi:10.4088/
JCP.18ac12473
14. Vimalanathan A, Gidyk DC, Diwan M, Gouveia
FV, Lipsman N, Giacobbe P, et al. Endocannabinoid
modulating drugs improve anxiety but not the expression
of conditioned fear in a rodent model of post-traumatic
stress disorder. Neuropharmacology. 2020;166:107965.
doi:10.1016/j.neuropharm.2020.107965
15. Botsford SL, Yang S, George TP. Cannabis and
cannabinoids in mood and anxiety disorders: Impact on
illness onset and course, and assessment of therapeutic
potential. Am J Addict. 2020;29:9-26. doi:10.1111/
ajad.12963
16. Giacobbe J, Marrocu A, Di Benedetto MG, Pariante CM,
Borsini A. A systematic, integrative review of the eects
of the endocannabinoid system on inammation and
neurogenesis in animal models of aective disorders.
Brain Behav Immun. 2021;93:353-67. doi: 10.1016/j.
bbi.2020.12.024
17. Maroon J, Bost J. Review of the neurological benets
of phytocannabinoids. Surg Neurol Int. 2018;9:91.
doi:10.4103/sni.sni_45_18
18. Meissner H, Cascella M. Cannabidiol (CBD). In
StatPearls, StatPearls Publishing, United States of
America; 2020. Available from: https://www.ncbi.nlm.
nih.gov/books/NBK556048/
19. Chen J, Hou C, Chen X, Wang D, Yang P, He X, et al.
Protective eect of cannabidiol on hydrogen peroxide
induced apoptosis, inammation and oxidative stress
in nucleus pulposus cells. Mol Med Rep. 2016;14:2321-
7. doi:10.3892/mmr.2016.5513
20. Limebeer CL, Rock EM, Sharkey KA, Parker LA.
Nausea-induced 5-HT release in the interoceptive
insular cortex and regulation by monoacylglycerol
lipase (MAGL) inhibition and cannabidiol.
eNeuro. 2018;5(4):ENEURO.0256-18. doi:10.1523/
ENEURO.0256-18.2018
21. Davies C, Bhattacharyya S. Cannabidiol as a
potential treatment for psychosis. er Adv
Psychopharmacol. 2019;9:2045125319881916.
doi:10.1177/2045125319881916
22. Li H, Liu Y, Tian D, Tian L, Ju X, Qi L, et al. Overview
of cannabidiol (CBD) and its analogues: Structures,
biological activities, and neuroprotective mechanisms
in epilepsy and Alzheimer’s disease. Eur J Med Chem.
2020;192:112163. doi:10.1016/j.ejmech.2020.112163
23. Silvestro S, Mammana S, Cavalli E, Bramanti P, Mazzon
E. Use of cannabidiol in the treatment of epilepsy:
Ecacy and security in clinical trials. Molecules.
2019;14:1459. doi:10.3390/molecules24081459
24. Lattanzi S, Brigo F, Trinka E, Zaccara G, Striano P,
Giovane CD, et al. Adjunctive Cannabidiol in Patients
with Dravet Syndrome: A systematic review and meta-
analysis of ecacy and safety. CNS Drugs. 2020;34:229-
41. doi:10.1007/s40263-020-00708-6
25. Flandreau EI, Toth M. Animal models of PTSD: A
critical review. Curr Top Behav Neurosci. 2018;38:47-
68. doi:10.1007/7854_2016_65.
26. Deslauriers J, Toth M, Der-Avakian A, Risbrough VB.
Current status of animal models of posttraumatic
stress disorder: Behavioral and biological phenotypes,
and future challenges in improving translation. Biol
Psychiatry. 2018;83(10):895-907. doi: 10.1016/j.
biopsych.2017.11.019
27. Verbitsky A, Dopfel D, Zhang N. Rodent models of
post-traumatic stress disorder: behavioral assessment.
Transl Psychiatry. 2020; 6;10(1):132. doi: 10.1038/
s41398-020-0806-x
28. Kilkenny C, Browne W, Cuthill IC, Emerson M, Altman
DG. NC3Rs Reporting Guidelines Working Group.
Animal research: reporting in vivo experiments: the
ARRIVE guidelines. Br J Pharmacol. 2010;160:1577-9.
doi:10.1111/j.1476-5381.2010.00872.x
29. McGrath JC, Lilley E. Implementing guidelines on
reporting research using animals (ARRIVE etc.): new
requirements for publication in BJP. Br J Pharmacol.
2015;172:3189-93. doi:10.1111/bph.12955
30. Percie du Sert N, Hurst V, Ahluwalia A, Alam S,
Avey MT, Baker M, et al. e ARRIVE guidelines 2.0:
Updated guidelines for reporting animal research. Br J
Pharmacol. 2020;177:3617-24. doi:10.1111/bph.15193
31. Kao CY, He Z, Zannas AS, Hahn O, Kühne C, Reichel JM,
et al. Fluoxetine treatment prevents the inammatory
response in a mouse model of posttraumatic stress
disorder. J Psychiatr Res. 2016;76:74-83. doi:10.1016/j.
jpsychires.2016.02.003
32. Ariel L, Inbar S, Edut S, Richter-Levin G. Fluoxetine
treatment is eective in a rat model of childhood-
induced post-traumatic stress disorder. Transl
Psychiatry. 2017;30;7(11):1260. doi: 10.1038/s41398-
017-0014-5
33. van der Kolk BA, Dreyfuss D, Michaels M, Shera D,
Berkowitz R, Fisler R, et al. Fluoxetine in posttraumatic
stress disorder. J Clin Psychiatry. 1994;55(12):517-22.
34. Martenyi F, Brown EB, Zhang H, Prakash A, Koke
SC. Fluoxetine versus placebo in posttraumatic stress
disorder. J Clin Psychiatry. 2002;63(3):199-206. doi:
10.4088/jcp.v63n0305
35. Long LE, Chesworth R, Huang XF, McGregor IS, Arnold
JC, Karl T. A behavioral comparison of acute and
chronic Delta9-tetrahydrocannabinol and cannabidiol
Cannabis Oil for Mood Disorders
Pharmaceutical Sciences, 2022, 28(2), 239-250 | 249
in C57BL/6JArc mice. Int J Neuropsychopharmacol.
2010;13:861-76. doi:10.1017/S1461145709990605
36. Moretti M, Budni J, Dos Santos DB, Antunes A,
Daufenbach JF, Manosso LM, et al. Protective eects
of ascorbic acid on behavior and oxidative status of
restraint-stressed mice. J Mol Neurosci. 2013;49:68-79.
doi:10.1007/s12031-012-9892-4
37. Schiavon AP, Bonato JM, Milani H, Guimarães FS,
Weort de Oliveira RM. Inuence of single and repeated
cannabidiol administration on emotional behavior
and markers of cell proliferation and neurogenesis
in non-stressed mice. Prog Neuropsychopharmacol
Biol Psychiatry. 2016;64:27-34. doi:10.1016/j.
pnpbp.2015.06.017
38. Freitas AE, Bettio LE, Neis VB. Santos DB, Ribeiro
CM, Rosa PB, et al. Agmatine abolishes restraint
stress-induced depressive-like behavior and
hippocampal antioxidant imbalance in mice. Prog
Neuropsychopharmacol Biol Psychiatry. 2014;50:143-
50. doi:10.1016/j.pnpbp.2013.12.012
39. Gáll Z, Farkas S, Albert Á, Ferencz E, Vancea S, Urkon
M, et al. Eects of chronic cannabidiol treatment in
the rat chronic unpredictable mild stress model of
depression. Biomolecules. 2020;10:801. doi:10.3390/
biom10050801
40. Poleszak E, Wlaź P, Kedzierska E, Nieoczym D, Wyska
E, Szymura-Oleksiak J, et al. Immobility stress induces
depression-like behavior in the forced swim test in
mice: eect of magnesium and imipramine. Pharmacol
Rep. 2006;58:746-52.
41. Budni J, Zomkowski AD, Engel D, Santos DB, Santos
AA, Moretti M, et al. Folic acid prevents depressive-
like behavior and hippocampal antioxidant imbalance
induced by restraint stress in mice. Exp Neurol.
2013;240:112-21. doi:10.1016/j.expneurol.2012.10.024
42. Lu XY, Kim CS, Frazer A, Zhang W. Leptin: a potential
novel antidepressant. Proc Natl Acad Sci U S A.
2006;103:1593-8. doi:10.1073/pnas.0508901103
43. Moretti M, Colla A, Balen GO, Santos DB, Bufni J,
Freitas AE, et al. Ascorbic acid treatment, similarly to
uoxetine, reverses depressive-like behavior and brain
oxidative damage induced by chronic unpredictable
stress. J Psychiatr Res. 2012;46:331-40. doi:10.1016/j.
jpsychires.2011.11.009
44. Porsolt RD, Le Pichon M, Jalfre M. Depression: a new
animal model sensitive to antidepressant treatments.
Nature. 1977;266:730-2. doi:10.1038/266730a0
45. Slattery DA, Cryan JF. Using the rat forced swim test
to assess antidepressant-like activity in rodents. Nat
Protoc. 2012;7:1009-14. doi:10.1038/nprot.2012.044
46. Quintans-Júnior LJ, Oliveira MG, Santana MF, Santana
MT, Guimarães AG, Siqueira JS, et al. α-Terpineol
reduces nociceptive behavior in mice. Pharm Biol.
2011;49:583-6. doi:10.3109/13880209.2010.529616
47. Diaz SL, Narboux-Nême N, Boutourlinsky K, Doly
S, Maroteaux L. Mice lacking the serotonin 5-HT2B
receptor as an animal model of resistance to selective
serotonin reuptake inhibitors antidepressants. Eur
Neuropsychopharmacol. 2016;26:265-79. doi:10.1016/j.
euroneuro.2015.12.012
48. Sulakhiya K, Patel VK, Saxena R, Dashore J, Srivastava
AK, Rathore M. Eect of Beta vulgaris Linn. leaves
extract on anxiety- and depressive-like behavior and
oxidative stress in mice aer acute restraint stress.
Pharmacognosy Res. 2016;8(1):1-7. doi:10.4103/0974-
8490.171100
49. Machado DG, Cunha MP, Neis VB, Balen GO,
Colla A, Grando J, et al. Fluoxetine reverses
depressive-like behaviors and increases hippocampal
acetylcholinesterase activity induced by olfactory
bulbectomy. Pharmacol Biochem Behav. 2012;103:220-
9. doi:10.1016/j.pbb.2012.08.024
50. Metna-Laurent M, Mondésir M, Grel A, Vallée M,
Piazza PV. Cannabinoid-Induced Tetrad in Mice. Curr
Protoc Neurosci. 2017;80:9.59.1-9.59.10. doi:10.1002/
cpns.31
51. Vera G, Cabezos PA, Martín MI, Abalo R.
Characterization of cannabinoid-induced relief of
neuropathic pain in a rat model of cisplatin-induced
neuropathy. Pharmacol Biochem Behav. 2013;105:205-
12. doi:10.1016/j.pbb.2013.02.008
52. Abizaid A, Merali Z, Anisman H. Cannabis: A potential
ecacious intervention for PTSD or simply snake
oil? J Psychiatry Neurosci. 2019;44:75-8. doi:10.1503/
jpn.190021
53. Vazhayil BK, Rajagopal SS, angavelu T,
Swaminathan G, Rajagounder E. Neuroprotective
eect of Clerodendrum serratum Linn. leaves extract
against acute restraint stress-induced depressive-like
behavioral symptoms in adult mice. Indian J Pharmacol.
2017;49:34-41. doi:10.4103/0253-7613.201028
54. Lezak KR, Missig G, Carlezon WA Jr. Behavioral
methods to study anxiety in rodents. Dialogues
Clin Neurosci. 2017;19:181-91. doi:10.31887/
DCNS.2017.19.2/wcarlezon
55. Flandreau EI, Toth M. Animal models of PTSD: A
critical review. Curr Top Behav Neurosci. 2018;38:47-
68. doi:10.1007/7854_2016_65
56. Zhu M, Shi J, Chen Y, Huang G, Zhu XW, Zhang
S, et al. Phosphodiesterase 2 inhibitor Hcyb1
reverses corticosterone-induced neurotoxicity and
depression-like behavior. Psychopharmacology (Berl).
2020;237:215-3224. doi:10.1007/s00213-019-05401-1
57. Resstel LB, Tavares RF, Lisboa SF, Joca SR, Corrêa FM,
Guimarães FS. 5-HT1A receptors are involved in the
cannabidiol-induced attenuation of behavioural and
cardiovascular responses to acute restraint stress in
rats. Br J Pharmacol. 2009;156:181-8. doi:10.1111/
j.1476-5381.2008.00046.x
58. Belardo C, Iannotta M, Boccella S, Rubino RC,
Ricciardi F, Infantino R, et al. Oral cannabidiol prevents
allodynia and neurological dysfunctions in a mouse
model of mild traumatic brain injury. Front Pharmacol.
2019;16:352. doi:10.3389/fphar.2019.00352.
Mendonça de Assis, et al.
250 | Pharmaceutical Sciences, 2022, 28(2), 239-250
59. Sales AJ, Crestani CC, Guimarães FS, Joca SRL.
Antidepressant-like eect induced by Cannabidiol
is dependent on brain serotonin levels. Prog
Neuropsychopharmacol Biol Psychiatry. 2018;86:255-
61. doi:10.1016/j.pnpbp.2018.06.002
60. Ferber SG, Namdar D, Hen-Shoval D, Eger G, Koltai
H, Shoval G, et al. e “entourage eect”: terpenes
coupled with cannabinoids for the treatment
of mood disorders and anxiety disorders. Curr
Neuropharmacol. 2020;18:87-96. doi:10.2174/157015
9X17666190903103923
61. McEwen BS, Eiland L, Hunter RG, Miller MM. Stress and
anxiety: structural plasticity and epigenetic regulation
as a consequence of stress. Neuropharmacology.
2012;62:3-12. doi:10.1016/j.neuropharm.2011.07.014
62. Monteiro S, Roque S, de Sá-Calçada D, Sousa N,
Correia-Neves M, Cerqueira JJ. An ecient chronic
unpredictable stress protocol to induce stress-related
responses in C57BL/6 mice. Front Psychiatry. 2015;6:6.
doi:10.3389/fpsyt.2015.00006
63. El-Alfy AT, Ivey K, Robinson K, Ahmed S, Radwan
M, Slade D, et al. Antidepressant-like eect of delta9-
tetrahydrocannabinol and other cannabinoids isolated
from Cannabis sativa L. Pharmacol Biochem Behav.
2010;95:434-42. doi:10.1016/j.pbb.2010.03.004
64. Schubart CD, Sommer IE, van Gastel WA, Goetgebuer
RL, Kahn RS, Boks MP. Cannabis with high cannabidiol
content is associated with fewer psychotic experiences.
Schizophr Res. 2011;130:216-21. doi:10.1016/j.
schres.2011.04.017
65. Tai S, Fantegrossi WE. Synthetic Cannabinoids:
Pharmacology, Behavioral Eects, and Abuse Potential.
Curr Addict Rep. 2014;1:129-136. doi:10.1007/s40429-
014-0014-y
66. Zagzoog A, Mohamed KA, Kim HJ, Kim ED, Frank
CS, Black T, et al. In vitro and in vivo pharmacological
activity of minor cannabinoids isolated from Cannabis
sativa. Sci Rep. 2020;10:20405. doi:10.1038/s41598-
020-77175-y
67. Linge R, Jiménez-Sánchez L, Campa L, Pilar-Cuéllar
F, Vidal R, Pazos A, et al. Cannabidiol induces rapid-
acting antidepressant-like eects and enhances cortical
5-HT/glutamate neurotransmission: role of 5-HT1A
receptors. Neuropharmacology. 2016;103:16-26.
doi:10.1016/j.neuropharm.2015.12.017
68. Bonaccorso S, Ricciardi A, Zangani C, Chiappini S,
Schifano F. Cannabidiol (CBD) use in psychiatric
disorders: A systematic review. Neurotoxicology.
2019;74:282-98. doi:10.1016/j.neuro.2019.08.002
69. Sarris J, Sinclair J, Karamacoska D, Davidson M, Firth
J. Medicinal cannabis for psychiatric disorders: a
clinically-focused systematic review. BMC Psychiatry.
2020;20:24. doi:10.1186/s12888-019-2409-8
70. Srivastava MD, Srivastava BI, Brouhard B. Delta9
tetrahydrocannabinol and cannabidiol alter
cytokine production by human immune cells.
Immunopharmacology. 1998;40:179-85. doi:10.1016/
s0162-3109(98)00041-1
71. Hegde VL, Singh UP, Nagarkatti PS, Nagarkatti M.
Critical role of mast cells and peroxisome proliferator-
activated receptor γ in the induction of myeloid-
derived suppressor cells by marijuana cannabidiol
in vivo. J Immunol. 2015;194:5211-22. doi:10.4049/
jimmunol.1401844
72. Vincent L, Vang D, Nguyen J, Benson B, Lei J, Gupta
K. Cannabinoid receptor-specic mechanisms to
alleviate pain in sickle cell anemia via inhibition of
mast cell activation and neurogenic inammation.
Haematologica. 2016;101:566-77. doi:10.3324/
haematol.2015.136523
73. Lindqvist D, Mellon SH, Dhabhar FS, Yehuda R,
Grenon SM, Flory JD, et al. Increased circulating blood
cell counts in combat-related PTSD: Associations
with inammation and PTSD severity. Psychiatry Res.
2017;258:330-6. doi: 10.1016/j.psychres.2017.08.052
74. Koraishy FM, Salas J, Neylan TC, Cohen BE, Schnurr
PP, Clouston S, et al. Association of severity of
posttraumatic stress disorder with inammation:
using total white blood cell count as a marker. Chronic
stress (thousand oaks). 2019;3:2470547019877651.
doi:10.1177/2470547019877651
75. Zhang H, Ding L, Shen T, Peng D. HMGB1 involved in
stress-induced depression and its neuroinammatory
priming role: a systematic review. Gen Psychiatr.
2019;32:e100084. doi:10.1136/gpsych-2019-100084
76. Franklin TC, Xu C, Duman RS. Depression and sterile
inammation: Essential role of danger associated
molecular patterns. Brain Behav Immun. 2018;72:2-13.
doi:10.1016/j.bbi.2017.10.025
77. Wang XW, Karki A, Du DY, Zhao XJ, Xiang XY, Lu ZQ.
Plasma levels of high mobility group box 1 increase in
patients with posttraumatic stress disorder aer severe
blunt chest trauma: a prospective cohort study. J Surg
Res. 2015;193:308-15. doi:10.1016/j.jss.2014.06.020
78. Apte RS, Chen DS, Ferrara N. VEGF in Signaling and
Disease: Beyond Discovery and Development. Cell.
2019;176:1248-64. doi:10.1016/j.cell.2019.01.021
79. Wheal AJ, Jadoon K, Randall MD, O’Sullivan SE. In
Vivo cannabidiol treatment improves endothelium-
dependent vasorelaxation in mesenteric arteries
of zucker diabetic fatty rats. Front Pharmacol.
2017;18(8):248. doi:10.3389/fphar.2017.00248.
80. Leishman E, Murphy M, Mackie K, Bradshaw HB.
Δ9-Tetrahydrocannabinol changes the brain lipidome
and transcriptome dierentially in the adolescent and
the adult. Biochim Biophys Acta Mol Cell Biol Lipids.
2018;1863:479-92. doi:10.1016/j.bbalip.2018.02.001
... With this in mind, biotechnological developments and extraction techniques have allowed the production of cannabis extracts and oils almost devoid of psychomimetic influence due to the isolation of the psychoactive substances [11]. Therefore, the broad-spectrum cannabis oil is nearly Δ9-THC free, but it contains all of the phytochemicals found in the plant, including terpenes, flavonoids, and other phytocannabinoids such as CBD [13]. Against this backdrop, the present study determined the broad-spectrum cannabis oil effects on reserpine-induced FM in mice, evaluating its analgesic effects using different routes of administration. ...
... The broad-spectrum cannabis oil was produced and analyzed by the Brazilian Association ABRACE (Associação Brasileira de Apoio Cannabis Esperança, Paraíba -Brazil/ National Register of Legal Entities -CNPJ under the number 23.877.015/0001-38). The chromatographic analysis was performed and published in a previous study and reported a CBD: Δ9-THC proportion of 11:1 and total cannabinoids of 40.2 % (15 mg of CBD in 1 ml of the oil) [13]. Regarding the microbiological assessment, the oil was under the current quality parameters [13]. ...
... The chromatographic analysis was performed and published in a previous study and reported a CBD: Δ9-THC proportion of 11:1 and total cannabinoids of 40.2 % (15 mg of CBD in 1 ml of the oil) [13]. Regarding the microbiological assessment, the oil was under the current quality parameters [13]. ...
Article
Full-text available
Fibromyalgia (FM) is an idiopathic disorder characterized by generalized pain and associated symptoms such as depression and anxiety. Cannabis sativa shows different pharmacological activities, such as analgesic, anti-inflammatory, neuroprotective, and immunomodulatory. Associated with this, the use of an oil with low concentrations of THC can reduce the psychomimetic adverse effects of the plant. Therefore, the present study aimed to evaluate the analgesic effect of broad-spectrum cannabis oil with low THC concentration in an experimental model of FM. Mechanical hyperalgesia, thermal allodynia, depressive- and anxious-related behavior, and locomotor activity were evaluated after reserpine (0.25 mg/kg; injected subcutaneously (s.c.) once daily for three consecutive days) administration. Our results showed that oral administration of broad-spectrum cannabis oil (0.1, 1, and 3 mg/kg, p.o.) in a single dose on the 4th day inhibited mechanical hyperalgesia and thermal allodynia induced by reserpine. Relevantly, treatment during four days with broad-spectrum cannabis oil (0.1 mg/kg, p.o.) reduced mechanical hyperalgesia 1 h after reserpine administration. Intraplantar treatment with cannabis oil significantly reversed mechanical and heat thermal nociception induced by reserpine injection. Interestingly, spinal and supraspinal administration of broad-spectrum cannabis oil completely inhibited mechanical hyperalgesia and thermal sensitivity induced by reserpine. The repeated cannabis oil administration, given daily for 14 days, markedly mitigated the mechanical and thermal sensitivity during the FM model, and its reduced depressive-like behavior induced by reserpine. In summary, broad-spectrum cannabis oil is an effective alternative to reverse the reserpine-induced fibromyalgia model.
Article
Full-text available
The endocannabinoid (eCB) system is considered relevant in the pathophysiology of affective disorders, and a potential therapeutic target, as its hypoactivity is considered an important risk factor of depression. However, the biological mechanisms whereby the eCB system affects mood remain elusive. Through a systematic review, thirty-seven articles were obtained from the PubMed/Medline, Web of Science, Embase, PsychInfo, and CINAHL databases, investigating the role of the eCB system on the immune system and neurogenesis, as well as resulting behavioural effects in rodent models of affective disorders. Overall, activation of the eCB system appears to decrease depressive-like behaviour and to be anti-inflammatory, while promoting neuro- and synaptogenesis in various models. Activation of cannabinoid receptors (CBRs) is shown to be crucial in improving depressive-like and anxiety-like behaviour, although cannabidiol administration suggests a role of additional mechanisms. CB1R signalling, as well as fatty acid amide hydrolase (FAAH) inhibition, are associated with decreased pro-inflammatory cytokines. Moreover, activation of CBRs is required for neurogenesis, which is also upregulated by FAAH inhibitors. This review is the first to assess the association between the eCB system, immune system and neurogenesis, alongside behavioural outcomes, across rodent models of affective disorders. We confirm the therapeutic potential of eCB system activation in depression and anxiety, highlighting immunoregulation as an important mechanism whereby dysfunctional behaviour and neurogenesis can be improved.
Article
Full-text available
The Cannabis sativa plant contains more than 120 cannabinoids. With the exceptions of ∆⁹-tetrahydrocannabinol (∆⁹-THC) and cannabidiol (CBD), comparatively little is known about the pharmacology of the less-abundant plant-derived (phyto) cannabinoids. The best-studied transducers of cannabinoid-dependent effects are type 1 and type 2 cannabinoid receptors (CB1R, CB2R). Partial agonism of CB1R by ∆⁹-THC is known to bring about the ‘high’ associated with Cannabis use, as well as the pain-, appetite-, and anxiety-modulating effects that are potentially therapeutic. CB2R activation by certain cannabinoids has been associated with anti-inflammatory activities. We assessed the activity of 8 phytocannabinoids at human CB1R, and CB2R in Chinese hamster ovary (CHO) cells stably expressing these receptors and in C57BL/6 mice in an attempt to better understand their pharmacodynamics. Specifically, ∆⁹-THC, ∆⁹-tetrahydrocannabinolic acid (∆⁹-THCa), ∆⁹-tetrahydrocannabivarin (THCV), CBD, cannabidiolic acid (CBDa), cannabidivarin (CBDV), cannabigerol (CBG), and cannabichromene (CBC) were evaluated. Compounds were assessed for their affinity to receptors, ability to inhibit cAMP accumulation, βarrestin2 recruitment, receptor selectivity, and ligand bias in cell culture; and cataleptic, hypothermic, anti-nociceptive, hypolocomotive, and anxiolytic effects in mice. Our data reveal partial agonist activity for many phytocannabinoids tested at CB1R and/or CB2R, as well as in vivo responses often associated with activation of CB1R. These data build on the growing body of literature showing cannabinoid receptor-dependent pharmacology for these less-abundant phytocannabinoids and are critical in understanding the complex and interactive pharmacology of Cannabis-derived molecules.
Article
Full-text available
Post-traumatic stress disorder (PTSD) is a well-known psychiatric disorder that affects millions of people worldwide. Pharmacodynamic and cognitive-behavioral therapies (CBT) have been used to treat patients with PTSD. However, it remains unclear whether there are concurrent changes in psychopathological and neurophysiological factors associated with PTSD patients. Past reports described those PTSD patients with efficient fatty acid metabolism, neurogenesis, mitochondrial energy balance could improve ability to cope against the conditioned fear responses and traumatic memories. Furthermore, cognitive, behavioral, cellular, and molecular evidence can be combined to create personalized therapies for PTSD sufferers either with or without comorbidities such as depression or memory impairment. Unfortunately, there is still evidence lacking to establish a full understanding of the underlying neurophysiological and psychopathological aspects associated with PTSD. This review has extensively discussed the single nucleotide polymorphism (SNPs) of genetic factors to cause PTSD, the implications of inflammation, neurotransmitter genomics, metabolic alterations, neuroendocrine disturbance (hypothalamus-pituitary-adrenal (HPA) axis), mitochondrial dynamics, neurogenesis, and premature aging related to PTSD-induced psychopathology and neurophysiology. In addition, the review delineated the importance of CBT and several pharmacodynamic therapies to mitigate symptomatology of PTSD.
Article
Full-text available
RationaleCurrently available PDE2 inhibitors have poor brain penetration that limits their therapeutic utility in the treatment of depression. Hcyb1 is a novel selective PDE2 inhibitor that was introduced more lipophilic groups with polar functionality to the scaffold pyrazolopyrimidinone to improve the blood-brain barrier (BBB) penetration. Our previous study suggested that Hcyb1 increased the neuronal cell viability and exhibited antidepressant-like effects, which were parallel to the currently available PDE2 inhibitor Bay 60-7550.Objectives The present study investigated whether Hcyb1 protected HT-22 cells against corticosterone-induced neurotoxicity and produced antidepressant-like effects in behavioral tests in stressed mice.Methods The neuroprotective effects of Hcyb1 against corticosterone-induced cell lesion were examined by cell viability (MTS) assay. The enzyme-linked immunosorbent assay (ELISA) and immunoblot analysis were used to determine the levels of cAMP or cGMP and expression of pCREB or BDNF, respectively, in the corticosterone-treated HT-22 cells. The antidepressant-like effects of Hcyb1 were determined in the tail suspension and novelty suppressed feeding tests in stressed mice.ResultsIn the cell-based assay, Hcyb1 significantly increased cell viability of HT-22 cells against corticosterone-induced neurotoxicity in a time- and dose-dependent manner. Hcyb1 also rescued corticosterone-induced decreases in both cGMP and cAMP levels, pCREB/CREB and BDNF expression. These protective effects of Hcyb1 were prevented by pretreatment with either the PKA inhibitor H89 or the PKG inhibitor KT5823. Moreover, Hcyb1 reversed acute stress–induced increases in immobility time and the latency to feed in the tail suspension and novelty suppressed feeding tests, respectively, which were prevented by pretreatment with H89 or KT5823.Conclusion These findings provide evidence that the neuroprotective effects of Hcyb1 are mediated by PDE2-dependent cAMP/cGMP signaling.
Article
Full-text available
Reproducible science requires transparent reporting. The ARRIVE guidelines (Animal Research: Reporting of In Vivo Experiments) were originally developed in 2010 to improve the reporting of animal research. They consist of a checklist of information to include in publications describing in vivo experiments to enable others to scrutinise the work adequately, evaluate its methodological rigour, and reproduce the methods and results. Despite considerable levels of endorsement by funders and journals over the years, adherence to the guidelines has been inconsistent, and the anticipated improvements in the quality of reporting in animal research publications have not been achieved. Here, we introduce ARRIVE 2.0. The guidelines have been updated and information reorganised to facilitate their use in practice. We used a Delphi exercise to prioritise and divide the items of the guidelines into 2 sets, the “ARRIVE Essential 10,” which constitutes the minimum requirement, and the “Recommended Set,” which describes the research context. This division facilitates improved reporting of animal research by supporting a stepwise approach to implementation. This helps journal editors and reviewers verify that the most important items are being reported in manuscripts. We have also developed the accompanying Explanation and Elaboration (E&E) document, which serves (1) to explain the rationale behind each item in the guidelines, (2) to clarify key concepts, and (3) to provide illustrative examples. We aim, through these changes, to help ensure that researchers, reviewers, and journal editors are better equipped to improve the rigour and transparency of the scientific process and thus reproducibility.
Article
Full-text available
Several neuropharmacological actions of cannabidiol (CBD) due to the modulation of the endocannabinoid system as well as direct serotonergic and gamma-aminobutyric acidergic actions have recently been identified. The current study aimed to reveal the effect of a long-term CBD treatment in the chronic unpredictable mild stress (CUMS) model of depression. Adult male Wistar rats (n = 24) were exposed to various stressors on a daily basis in order to induce anhedonia and anxiety-like behaviors. CBD (10 mg/kg body weight) was administered by daily intraperitoneal injections for 28 days (n = 12). The effects of the treatment were assessed on body weight, sucrose preference, and exploratory and anxiety-related behavior in the open field (OF) and elevated plus maze (EPM) tests. Hair corticosterone was also assayed by liquid chromatography–mass spectrometry. At the end of the experiment, CBD-treated rats showed a higher rate of body weight gain (5.94% vs. 0.67%) and sucrose preference compared to controls. A significant increase in vertical exploration and a trend of increase in distance traveled in the OF test were observed in the CBD-treated group compared to the vehicle-treated group. The EPM test did not reveal any differences between the groups. Hair corticosterone levels increased in the CBD-treated group, while they decreased in controls compared to baseline (+36.01% vs. −45.91%). In conclusion, CBD exerted a prohedonic effect in rats subjected to CUMS, demonstrated by the increased sucrose preference after three weeks of treatment. The reversal of the effect of CUMS on hair corticosterone concentrations might also point toward an anxiolytic or antidepressant-like effect of CBD, but this needs further confirmation.
Article
Full-text available
Although the etiology and expression of psychiatric disorders are complex, mammals show biologically preserved behavioral and neurobiological responses to valent stimuli which underlie the use of rodent models of post-traumatic stress disorder (PTSD). PTSD is a complex phenotype that is difficult to model in rodents because it is diagnosed by patient interview and influenced by both environmental and genetic factors. However, given that PTSD results from traumatic experiences, rodent models can simulate stress induction and disorder development. By manipulating stress type, intensity, duration, and frequency, preclinical models reflect core PTSD phenotypes, measured through various behavioral assays. Paradigms precipitate the disorder by applying physical, social, and psychological stressors individually or in combination. This review discusses the methods used to trigger and evaluate PTSD-like phenotypes. It highlights studies employing each stress model and evaluates their translational efficacies against DSM-5, validity criteria, and criteria proposed by Yehuda and Antelman’s commentary in 1993. This is intended to aid in paradigm selection by informing readers about rodent models, their benefits to the clinical community, challenges associated with the translational models, and opportunities for future work. To inform PTSD model validity and relevance to human psychopathology, we propose that models incorporate behavioral test batteries, individual differences, sex differences, strain and stock differences, early life stress effects, biomarkers, stringent success criteria for drug development, Research Domain Criteria, technological advances, and cross-species comparisons. We conclude that, despite the challenges, animal studies will be pivotal to advances in understanding PTSD and the neurobiology of stress.
Article
Full-text available
Neuroimaging findings suggest that the amygdala plays a primary role in both the psychopathology of posttraumatic stress disorder (PTSD) and poor sleep quality, which are common in trauma survivors. However, the neural mechanisms of these two problems in trauma survivors associated with amygdala remain unclear. In the current study, we aimed to explore the role of functional connectivity of amygdala subregions in both PTSD symptoms and poor sleep quality. A total of 94 trauma-exposed subjects were scanned on a 3T MR system using resting-state functional magnetic resonance imaging. Both Pittsburgh Sleep Quality Index and Clinician-Administered PTSD Scale scores were negatively correlated with the resting-state functional connectivity between the left basolateral amygdala-left medial prefrontal cortex and the right basolateral amygdala-right medial prefrontal cortex. Our findings suggest a shared amygdala subregional neural circuitry underlying the neuropathological mechanisms of PTSD symptoms and poor sleep quality in trauma survivors.
Article
Background Dravet syndrome (DS) is one of the most severe forms of drug-resistant epilepsy and available interventions fail to control seizures in most patients. Cannabidiol (CBD) is the first in a new class of antiepileptic drugs with a distinctive chemical structure and mechanism of action. Objective The aim of this systematic review was to evaluate the efficacy and safety of CBD as adjunctive treatment for seizures in patients with DS using meta-analytical techniques. Methods We searched for randomized, placebo-controlled, single- or double-blinded trials. Main outcomes included ≥ 50% reduction in baseline convulsive seizure frequency and the incidence of treatment withdrawal and adverse events (AEs). Risk ratios (RRs) with 95% confidence intervals (95% CIs) were estimated through the inverse variance method. Results Three trials were included involving 359 participants, 228 for CBD and 131 for placebo groups. In all trials, the active treatment was a plant-derived pharmaceutical formulation of purified CBD oral solution. The pooled RR for 50% response during the treatment was 1.69 (95% CI 1.21–2.36; p = 0.002). Across the trials, treatment was discontinued in 20 (9.0%) and 3 (2.3%) cases in the add-on CBD and placebo groups, respectively; the RR for CBD withdrawal was 3.12 (95% CI 1.07–9.10; p = 0.037). The RR to develop any AE during add-on CBD treatment was 1.06 (95% CI 0.87–1.28; p = 0.561). AEs significantly associated with adjunctive CBD were somnolence, decreased appetite, diarrhea, and increased serum aminotransferases. Conclusions Adjunctive CBD resulted in a greater reduction in convulsive seizure frequency than placebo and a higher rate of AEs in patients with DS presenting with seizures uncontrolled by concomitant antiepileptic therapy.