You are on page 1of 11

Biomedicine & Pharmacotherapy 109 (2019) 1698–1708

Contents lists available at ScienceDirect

Biomedicine & Pharmacotherapy


journal homepage: www.elsevier.com/locate/biopha

Protective effect of Convolvulus pluricaulis against neuroinflammation T


associated depressive behavior induced by chronic unpredictable mild stress
in rat
⁎,1
Girdhari Lal Gupta , Joneth Fernandes1
Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai, 400 056, India

ARTICLE INFO ABSTRACT

Keywords: Depression is a heterogeneous disorder and has been regarded as an inflammatory disease. The aerial parts of the
Chronic unpredictable mild stress Convolvulus pluricaulis are used in Indian traditional medicines for the management of nervous disorders.
Convolvulus pluricaulis However, the influence of methanolic extract of aerial parts of Convolvulus pluricaulis (CPE) on a chronic animal
Depression model of depression has not been investigated yet, and associated biochemical changes are still unclear.
Cytokines
Therefore, this study investigates the effects of CPE on a chronic rat model of depression and explores its un-
Interleukin-1β
Interleukin-6
derlying mechanism of action on neuroinflammation and brain monoamines. The antidepressant-like effect of
Tumor necrosis factor-α CPE (25, 50, and 100 mg/kg, p.o.) was depicted using the sucrose preference test and the forced swimming test
ALT (FST) while CUMS-induced alteration in the locomotor index was measured using the open field test (OFT) and
AST actophotometer. A consecutive one-week treatment of CPE (50, and 100 mg/kg) or fluoxetine (10 mg/kg, p.o.)
ALP treatment significantly increased sucrose preference index, reduced immobility time in the FST, and increased
the number of squares crossed, the number of rearing in the OFT and locomotion in the actophotometer in the
CUMS-exposed rats. Moreover, elevated levels of pro-inflammatory cytokines IL-1β, IL-6, TNF-α and liver bio-
markers ALT, AST were also significantly reversed by CPE (50, and 100 mg/kg) or fluoxetine administration in
the CUMS-exposed rats. Furthermore, a one-week treatment of CPE (50 and 100 mg/kg) or fluoxetine also re-
markably restored the serotonin and noradrenaline levels in the hippocampus as well as in the prefrontal cortex
of the CUMS-exposed rats. However, CPE (25 mg/kg) exerted insignificant protection against CUMS-induced
depressive-like behavior and associated neuroinflammation. Therefore, this study demonstrates that CPE exerted
antidepressant-like effect which could be mediated by anti-inflammatory potential, restoring liver biomarkers or
monoaminergic responses in the stressed rats.

1. Introduction depression, most of the current antidepressants used in the clinical


practice not only have a low efficacy, but they also may cause side
As stated by the World Health Organization, depression affects an effects [8–10]. Therefore, the development of safer and effective newer
estimate of 300 million people of all ages globally and is a highly de- medications is always desired world-wide. Chiefly, antidepressants
bilitating, life-threatening psychiatric illness [1,2]. Depression is re- based on the monoaminergic deficiency hypothesis have a key role in
garded to become the second most disabling burden in the World by the clinical practice but are also associated to depict treatment-resistant
2030 [3,4]. Depression leads to pathological stages like anhedonia, in one-third of the patients [11–13]. However, the current focus is also
helplessness feelings, reduced physical activity, negative moods, weight on treatment strategies that influence the distinct neurochemical sys-
loss, low morale, and cognitive dysfunctions, etc. [5–7]. Despite en- tems [14]. Increasing evidence has claimed that depression is an oxi-
ormous progress have been made to understand the pathogenesis of dative and inflammatory disorder [15–19]. Clinical evidence also

Abbreviations: CUMS, chronic unpredictable mild stress; ELISA, enzyme-linked immunosorbent assay; SPT, sucrose preference test; OFT, open field test; FST, forced
swimming test; HPTLC, high-performance thin layer chromatography; IL-1β, interleukin-1beta; IL-6, interleukin-6; TNF-α, tumor necrosis factor-α; ALT, Alanine
transaminase; AST, Aspartate transaminase; ALP, Alkaline phosphatase

Corresponding author at: Department of Pharmacology, School of Pharmacy & Technology Management, SVKM’s NMIMS, V. L. Mehta Road, Vile Parle (W),
Mumbai, 400 056, India.
E-mail address: girdhari_gupta@rediffmail.com (G.L. Gupta).
1
Both authors contributed equally to this work.

https://doi.org/10.1016/j.biopha.2018.11.046
Received 20 October 2018; Received in revised form 3 November 2018; Accepted 10 November 2018
0753-3322/ © 2018 The Authors. Published by Elsevier Masson SAS. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/BY-NC-ND/4.0/).
G.L. Gupta, J. Fernandes Biomedicine & Pharmacotherapy 109 (2019) 1698–1708

closely associates inflammation with the complicated pathogenesis of Laboratories Pvt. Ltd., Mumbai, India), norepinephrine (NE) (Sigma-
neuropsychiatric disorders, which include depression [4,20]. Several Aldrich, St. Louis, MO, United States), and fluoxetine (Sun Pharma,
reports have revealed the close association of the expression of pro- India) were procured from the respective sources. Alanine transaminase
inflammatory mediators and depression-like behavior [16,20,21]. (ALT) kit, aspartate transaminase (AST) kit, and alkaline phosphatase
Moreover, pro-inflammatory cytokines including interleukin-1β (IL-1 (ALP) kit were purchased from ERBA Diagnostics, Mannheim. HPTLC
β), interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) have been grade methanol was purchased from S.D. Fine Chemical Ltd. (Mumbai,
engrossed to increase during depression in animals [4,17,19,21]. De- India). All other chemicals and reagents were of analytical grades and
pressive patients have also depicted monoaminergic deficiency and were purchased from Sisco Research Laboratories Pvt. Ltd., Mumbai,
elevated levels of pro-inflammatory cytokines in the peripheral circu- India, and Merk India Ltd., Mumbai, India. Commercial kits of Rat IL-1β
lation and some brain regions [20,22,23]. Although the pathogenesis of ELISA, Rat IL-6 ELISA, and Rat TNFα ELISA were purchased from
depression progression has been elucidated extensively, further studies Krishgen Biosystems, India. Convolvulus pluricaulis and fluoxetine were
are still needed to assimilate the relevant molecular mechanism. Ad- dissolved in 0.5% w/v of sodium–carboxymethylcellulose (Na-CMC) in
ditionally, most of the reports on depression primarily focus on changes distilled water. Oral administration of CPE and fluoxetine were selected
in the brain areas including the hippocampus and prefrontal cortex and considered as the most preferred route of administration in the
[24–26]. psychiatric patients.
Herbal medicines are gaining attention for the development of new
therapeutically active compounds with higher potency and lower 2.2. Animals and housing
toxicity. A perennial herb Convolvulus pluricaulis Choisy
(Convolvulaceae) commonly known as Shankpushpi has been reported Male Wistar rats (7–8 weeks, weighing 120–150 g) were supplied by
to depict multiple pharmacological activities, including improvement of Bharat Serum and Vaccines Ltd, Thane, India, and housed in a con-
learning and memory in young and old mice [27] and clinically in trolled environment with 25 ± 2 °C temperature, 70 ± 10% relative
improving memory, especially in long-term memory loss in the younger humidity and 12 h light/12 h dark cycle in well-ventilated poly-
age group [28]. Convolvulus pluricaulis has ameliorated human micro- propylene cages. Standard commercial rodent pellets (Nutrimix
tubule-associated protein tau-induced neurotoxicity in Alzheimer's Laboratory Animal Feed, Maharashtra, India) and water were provided
disease in Drosophila model [29], declined tau and amyloid precursor ad libitum. The rats were acclimatized to the laboratory conditions for 7
protein expression [30], prevented impairment of cholinergic and an- days before the experiments. An approval prior to animal experi-
tioxidant activity induced by aluminium in rat brain cerebral cortex mentation was obtained from the Institutional Animal Ethics
[31], depicted neuroprotective activity [32,33], attenuated scopola- Committee, constituted as per Committee for the Purpose of Control
mine-induced amnesia [34], exhibited neuroprotective activity against and Supervision of Experiments on Animals (CPCSEA), Govt. of India
tumor necrosis factor-alpha (TNF-α) and prostaglandin E2 [33], and (CPCSEA/IAEC/P-93/2017).
reduced 3- nitropropionic acid-induced neurotoxicity in rats [35]. Ad-
ditionally, Convolvulus pluricaulis also attenuated alcohol addiction in 2.3. Plant material and preparation of the methanolic extract
mice [36], elicited a significant antidepressant-like effect [37], and
decreased Triton WR-1339-induced hyperlipidemia [38]. Convolvulus Dried aerial parts of Convolvulus pluricaulis was purchased from
pluricaulis is also known for demonstrating antioxidant, anticonvulsant Konark Herbals, Mumbai, India. The authentication of the plant
activity [39,40], anxiolytic effects [41], and antiulcerogenic effect [42]. Convolvulus pluricaulis was carried out by scientists of
Moreover, the plant is mentioned as one of the best Medhya Rasayana Agharkar Research Institute, grant-in-aid research institute of the
(nervine tonic) in Ayurveda, and the aerial parts of the plant are used Department of Science and Technology (DST), Government of India,
locally in India for hundreds of years for various nervous disorders. This Pune, India. The voucher specimen (no. 1402/2018) was kept for the
herb is also marketed as a single or multi-herb formulation. The active future reference. The plant was coarsely powdered with a mechanical
constituents of the herb include alkaloid (shankhapushpine), flavonoids grinder and passed through sieve number 60 and stored in a sealed
(kaempferol derivatives), phytosterol (β-sitosterol), 20-oxodo- container. The coarsely powdered plant material was defatted with n-
triacontanol, tetra-triacontanoic acid, 29-oxodotriacontanol and sco- hexane. The defatted, air-dried plant powder was further extracted in
poletin [33,35,43,44]. Interestingly, scopoletin has been reported to soxhlet apparatus with methanol at 50 °C for 48 h. The use of metha-
depict antidepressant-like effect through monoaminergic systems [45] nolic solvent has been reported to be rich in alkaloids and coumarins
and also suppresses pro-inflammatory cytokines [46]. [38,48]. The solvent was recovered using rotary evaporator (Heidolph,
Chronic unpredictable mild stress (CUMS) consists of mild stressors Germany) under reduced pressure of 600 mmHg at 40 °C. The semisolid
which imitate the common stressors in routine human life. It results in mass was further lyophilized for 24 h. The methanolic extraction of dry
alterations in neural, endocrine variables and behavioral changes [47]. aerial plant material (1 kg) yielded 81 g (8.1% w/w) of the extract. The
Moreover, the influence of Convolvulus pluricaulis on a chronic animal lyophilized dry powder was sealed in amber bottles and stored in a 4 °C
model of depression has not been elucidated yet, and associated bio- freezer before use.
chemical changes, an inflammatory link is still unclear. Therefore, the
present investigation was undertaken to confirm the antidepressant-like 2.4. Phytochemical analysis
effects of the methanolic extract of the aerial parts of Convolvulus
pluricaulis extract (CPE) in the CUMS animal model and to investigate Preliminary phytochemical screening of CPE was carried out qua-
whether the underlying mechanism accountable for the beneficial ef- litatively for the presence of carbohydrates, proteins, amino acids, al-
fects of CPE are associated with alterations in pro-inflammatory bio- kaloids, glycosides, flavonoids, saponins, and tannins by utilizing
markers in plasma and monoamines in the hippocampus and prefrontal standard procedures [48,49].
cortex regions of the brain.
2.5. Quantification of scopoletin in CPE by High-performance thin layer
2. Materials and methods chromatography (HPTLC) method

2.1. Drugs doses and routes of administration The reference compound scopoletin was quantitated in CPE as re-
ported in the monograph of Convolvulus pluricaulis in Quality Standards
Convolvulus pluricaulis (Konark Herbals, Mumbai), serotonin (Tokyo of Indian Medicinal Plants published by Indian Council of Medicinal
Chemical Industry Co., Ltd., Japan), scopoletin (Sisco Research Research, Government of India, New Delhi, India [49] with slight

1699
G.L. Gupta, J. Fernandes Biomedicine & Pharmacotherapy 109 (2019) 1698–1708

euthanized and their vital organs were individually depicted for mac-
roscopic evaluation.

2.7. Experimental design

All behavioral studies were executed between 09:00 and 15:00 h in


the naive experimental rats. After acclimatization to the laboratory
conditions, 36 rats were randomly divided into six groups: (1) Non-
stressed + vehicle (control); (2) CUMS + vehicle; (3–5) three CPE-
treated groups (25, 50, and 100 mg/kg of CPE + CUMS); and (6)
CUMS + 10 mg/kg, p.o. of standard drug fluoxetine. The doses of CPE
and fluoxetine were selected based on the previous pharmacological
reports [35,37,51–53]. CPE and fluoxetine were administrated once a
day for the last seven days of the CUMS exposure (from day 21 to day
27). The rats in the normal control group and CUMS group received an
equivalent volume of 0.5% Na-CMC (10 ml/kg, p.o., once a day, from
day 21 to day 27). One-hour after the drug administration, behavioral
Fig. 1. High-performance thin layer chromatography fingerprints of (A) stan- tests were executed, and subsequently animals were sacrificed for fur-
dard scopoletin (Rf 0.67) and (B,C,D) scopoletin in the methanolic CPE in tri- ther biochemical determinations. The detailed schedule of the experi-
plicate using pre-coated silica gel aluminum plate 60 F254, mobile phase ethyl mental illustration is presented in Table 1.
acetate: toluene: acetic acid, 5: 4: 1, v/v/v at 300 nm.
2.8. Chronic unpredictable mild stress procedure
modification using HPTLC (GmbH/SARSTEDT; Desaga, Germany), in-
cluding AS30 HPTLC applicator, CD60 HPTLC densitometer with Pro Rats were exposed to a variety of mild environmental or psycho-
Quant Windows software. Briefly, CPE was dissolved in methanol at a social stressors as described previously [54,55] with minor modifica-
concentration of 20 mg/ml and scopoletin at 2 mg/mL. The calibration tions as per the schedule in Table 2. Briefly, the control group rats were
curve for scopoletin was prepared on a pre-coated silica gel aluminum housed with 3 rats per cage with free access to food, and water and
plate 60 F254 of 0.2 mm thickness (E. Merck, Germany). For the esti- were left unstressed in their home cages with the exception of general
mation of scopoletin in the extract, one band of the standard scopoletin cleaning and handling. However, rats in the CUMS and drug-treated
solution of volume 20 μl and CPE test solution of volume 20 μl was groups were isolated in the individual cages and exposed to the fol-
applied in triplicate on a precoated TLC plate. Glass twin-trough lowing nine stressors for 27 days: (1) Warm swimming at 40 °C for
chamber saturated with the mobile phase was used for the development 5 min; (2) 24 h of water deprivation; (3) 24 h of the cage tilting (30°);
of TLC plate to a height of 80 mm. The mobile phase was comprised of (4) tail pinching for 1 min (1 cm from the tip of the tail); (5) 24 h of food
ethyl acetate: toluene: acetic acid (5: 4: 1 v/v/v). The plates were air deprivation; (6) overnight illumination; (7) overhang (10 min); (8) cold
dried and scanned at 300 nm. The peak area under the curve was re- swimming at 8 °C for 5 min; (9) physical restraint for 2 h. These stres-
corded, and a calibration curve for scopoletin was plotted. The amount sors were executed once daily between 09:00 h and 13:00 h (except the
of scopoletin present in the samples was calculated from the calibration stressors of 24 h) with a variable unpredictable sequence for con-
curve. Photo documentation of scopoletin is presented in Fig. 1. secutive 27 days and each stressor was repeated three times within 27
days. Prophesy and adjustment to any particular stressor were arrested
by not repeating the same stressor for the two consecutive days.
2.6. Acute toxicity study
2.9. Behavioral tests
An acute toxicity study was conducted as per the guidelines of the
Organization for Economic Co-Operation and Development 423 [50]. After the termination of the CUMS procedure, trails were executed
Female Wistar rats were fasted overnight and administered a single in a sound-attenuated room using the standard behavior paradigm in
dose of 0.5% Na-CMC (control group) (n = 3) or 2000 mg/kg of CPE the order listed in Table 1, i.e., open field test (OFT) on day 28, acto-
dissolved in 0.5% Na-CMC (n = 3) orally. Thereafter, the rats were photometer test on day 29, sucrose preference test (SPT) on day 30 and
monitored continuously for 14 days. On the last 14 day, the rats were 31, and forced swim test (FST) on day 33 and 34. Only one test was

Table 1
Timeline of the experimental design.
Day (-7 to 0) Day 1 - Day Day 21 - Day 27 Day 28 - Day 34 Day 35
20

Adaptation to Lab CUMS exposure (Except normal control group) Behavioral tests Sacrifice and sample collection
(Not counted in the Drug Treatment (10 ml/kg, p.o.
study) administration, once daily)
Non-CUMS Normal control group: vehicle OFT (on day 28) Estimation of serotonin and noradrenaline levels in the
hippocampus and prefrontal cortex
CUMS CUMS group: vehicle Actophotometer (on day 29)
CPE (25 mg/kg) Estimation of pro-inflammatory cytokines (IL-1β, IL-6,
CPE (50 mg/kg) SPT (on day 30 and 31) and TNF-α) and
CPE (100 mg/kg) liver biomarkers (ALT, AST, and ALP) in plasma
Fluoxetine (10 mg/kg) FST (on day 33 and 34)

CUMS, chronic unpredictable mild stress; CPE, methanolic extract of areal part of Convolvulus pluricaulis; OFT, open field test; SPT, sucrose preference test; FST,
forced swimming test; IL-1β, interleukin-1β; IL-6, interleukin-6; TNF-α, tumor necrosis factor-α; ALT, Alanine transaminase; AST, Aspartate transaminase; ALP,
Alkaline phosphatase.

1700
G.L. Gupta, J. Fernandes Biomedicine & Pharmacotherapy 109 (2019) 1698–1708

Table 2 instructions. The mean absorbance was determined for each set of du-
The stressors of chronic unpredictable mild stress procedure with specified plicate standards and samples. The levels of cytokines IL-1β, IL-6, and
days. TNFα were expressed as pg/mL. For biochemical estimation, blood was
Stressors Days of CUMS exposure collected without using an anticoagulant and allowed to clot for 30 min
at 25 °C. Thereafter, centrifuged at 1500×g for 15 min at 4 °C to collect
Warm swimming at 40 °C for 5 min 1 16 25 the serum, which was stored at −20 °C. The level of liver enzymes ALT,
24 h of water deprivation 2 15 21
AST and ALP were measured using commercially available standard
24 h of cage tilting (30°) 3 12 24
Tail pinching for 1 min (1 cm from the tip of the 4 10 23 biochemical assay kit. Post-blood withdrawal on day 35, rats were sa-
tail) crificed by decapitation to remove the brain and isolated brain tissues
24 h of food deprivation 5 11 22 i.e. the hippocampus and prefrontal cortex. The brain tissues were
Overnight illumination 6 14 20 frozen (−80 °C) for further analysis of serotonin and noradrenaline
Overhang (10 min) 7 13 19
Cold swimming at 8 °C for 5 min 8 17 26
levels in the prefrontal cortex and the hippocampus.
Physical restraint for 2 h 9 18 27
2.11. HPLC analysis of brain tissue homogenate to measure serotonin levels

performed each day keeping a 24 h lapse between the sucrose pre- The prefrontal cortex and hippocampus were homogenized in 2 ml
ference test and the forced swim test. The behavioral trials were per- of 0.1 N perchloric acid and centrifuged at 8000×g for 20 min at 4 °C
formed by a trained observer who was unaware of the experimental separately. The supernatant of the tissue homogenate was collected and
groups. The use of fluoxetine as a positive control was exerted because stored at −80 °C until use. This supernatant was further filtered using a
of its routine use in the treatment of CUMS-induced depression-like 0.45 μm membrane filter (Durapore, Millipore) and tested for serotonin
behavior [56]. concentration by using the HPLC method [62]. Chromatography was
performed with HPLC (SHIMADZU: LC-2010 CHT, Shimadzu, Kyoto,
2.9.1. Open-field test Japan), equipped with 25 cm length, 4.6 mm internal diameter Kro-
The OFT was executed under bright ambient room light to assess the masil C18 Column (particle size 5 mm) (Kromasil CA). The clear tissue
locomotor and exploratory behavior of the rats as previously reported homogenate was injected in triplicates and chromatographic separation
[55]. The rats were placed individually into the center area and allowed was achieved at 280 nm using 0.05% formic acid and acetonitrile
to explore the unfamiliar arena for 5 min. The number of total squares (90:10 v/v) as the mobile phase with 1 ml/min flow rate. Standard
crossed, number of rearing (number of times the rat raised on its hind serotonin weighing 10 mg was diluted to 10 ml with the mobile phase
legs), and grooming episodes (washing of the coat) were registered. to make a 1000 μg/ml stock solution. The stock solution was further
Post-exposure of each rat, the OFT apparatus was wiped with a 50% diluted with the mobile phase to get 100, 200, 300 and 400 μg/ml so-
ethanol to abolish any sign of olfactory cues. lutions. These four dilutions were scanned in triplicates at 280 nm, and
the chromatographs were recorded to obtain the retention time and
2.9.2. Spontaneous locomotor activity peak area under the curve. The calibration curve was obtained by
Locomotion of each rat was investigated in the actophotometer plotting the peak area under the curve versus concentration. Ad-
(Dolphin) for the period of 5 min [57]. ditionally, CPE and fluoxetine-treated rat brain tissue homogenate
samples were scanned in triplicates at 280 nm. Finally, the concentra-
2.9.3. Sucrose preference test tion of serotonin was calculated using the calibration curve.
The SPT was used to observe anhedonic-like behavior and executed
as reported earlier with minor modifications [47,58]. Initially, rats 2.12. Statistical analysis
were trained to consume 2% w/v sucrose solution for 24-hours by
placing two bottles of sucrose solution in each cage to prevent neo- The data were statistically analyzed using GraphPad Prism Software
phobia-induced changes in the preference. After a 12-hour period of version 7.00, San Diego, California, USA, and expressed as a mean ±
food and water deprivation, all rats were given free access to two standard error of the mean (S.E.M.) (n = 6). Data were analyzed by
standard drinking bottles, one containing 2.5% sucrose and the other two-way analysis of variance (ANOVA), followed by post-hoc Tukey’s
with tap water for the period of three hours. The sucrose and water multiple comparison tests. Differences with p < 0.05 were considered
intake were measured, and sucrose preference is presented as the per- statistically significant.
centage of total volume of fluid consumed.
Volume of sucrose solution ingested 3. Results
Sucrose preference % = x 100
Volume of total liquid ingested
3.1. Phytochemical analysis of CPE

2.9.4. Forced swim test Preliminary phytochemical screening of CPE depicted the presence
To ascertain the helplessness characteristic of the depression, fre- of carbohydrates, proteins, amino acids, alkaloids, glycosides, flavo-
quently used FST model was standardized in our laboratory [59–61]. noids and, saponins. The HPTLC analysis depicted well-resolved peaks
Rats were subjected singly in a transparent cylinder and the immobility of CPE revealing the presence of scopoletin. The calibration curve for
time was recorded for the period of 5 min. scopoletin was linear in the range of 1.0–5.0 μg/mL. The linear re-
gression equation for the calibration curve was y = 57.087x+699.34;
2.10. Blood sampling and tissue extraction (R2 = 0.9907) where y is the peak area response at 300 nm and x re-
present the concentration in μg/mL. The spots of the entire chromato-
After the behavior tests on day 35, rats were anesthetized with gram were visualized under 300 nm and the percentage of scopoletin
pentobarbital sodium (45 mg/kg) by intraperitoneal injection, and (Rf 0.67) in CPE was found to be 0.106% w/w (Fig. 1).
blood was collected from the retro-orbital sinus. Ethylene-diamine-
tetraacetate coated vials were used to collect the blood sample for es- 3.2. Acute oral toxicity studies
timation of pro-inflammatory cytokine IL-1β, IL-6, and TNFα. The
plasma pro-inflammatory cytokine levels were detected in duplicate No mortality or morbidity was depicted in the rats during the ob-
with commercial ELISA kits according to the manufacturer's servation period (14 days) following a single administration of CPE

1701
G.L. Gupta, J. Fernandes Biomedicine & Pharmacotherapy 109 (2019) 1698–1708

Fig. 2. Effects of CUMS and CPE treatment on


the number of total squares crossed, number of
rearing, and number of grooming episodes in
the OFT. Results are shown as mean ± SEM
(n = 6) by vertical bars, where **p < 0.01,
****
p < 0.0001 (Compared with normal con-
trol group), #p < 0.05, ##p < 0.01,
###
p < 0.001, ####p < 0.0001 (Compared
with vehicle-treated CUMS-exposed rats), Two-
way analysis of variance (ANOVA), followed by
post-hoc Tukey’s multiple comparison test.
CUMS, chronic unpredictable mild stress; CPE,
methanolic extract of areal part of Convolvulus
pluricaulis; OFT, open field test.

Fig. 3. Effect of CUMS and CPE treatment on


the locomotor index in the actophotometer.
Results are shown as mean ± SEM (n = 6) by
vertical bars, where ****p < 0.0001
(Compared to normal control group), #p <
0.05, ###p < 0.001, ###p < 0.001
(Compared to vehicle-treated CUMS-exposed
rats), Two-way analysis of variance (ANOVA),
followed by post-hoc Tukey’s multiple com-
parison test. CUMS, chronic unpredictable mild
stress; CPE, methanolic extract of areal part of
Convolvulus pluricaulis.

(2000 mg/kg, p.o.). Morphological features (eyes, nose, fur, and skin) differences in the numbers of grooming among all CPE and fluoxetine-
appeared normal. No tremors, diarrhea, convulsions, salivation, le- treated groups (p > 0.05; Fig. 2) in the CUMS as well as normal control
thargy or unusual behavior like walking backward, self-mutilation were rats. Additionally, the separate experiment was executed to confirm the
monitored. There were no macroscopic alterations in the brain, liver, drugs escalating locomotor index which may endanger false-positive
heart, kidney, and stomach (data presented in a supplementary sheet as behavioral results. In that study, rats were unexposed to the CUMS
Fig. 1S). This indicates that CPE was safe up to a single dose of procedure (normal rats), and one-week CPE administration (25, 50, and
2000 mg/kg, p.o. in experimental rats, and the extract was classified as 100 mg/kg, p.o.) did not alter the total number of squares crossed (p >
safe (category 5), according to the OECD 423 guidelines. 0.05), the number of rearing (p > 0.05) and number of grooming (p >
0.05) in the OFT and locomotor score in the actophotometer (p > 0.05)
when compared with vehicle-treated normal control group (Data pre-
3.3. Effects of CUMS and CPE in the open field test sented as supporting information, Fig. 2S & 3S). Therefore, this clearly
indicates that elevated locomotion in the OFT and reduced immobility
As illustrated in Fig. 2, reduction in the number of squares crossed in the FST on the CPE treatment in the CUMS-exposed rats cannot be
and the number of rearing were noticed significantly in the CUMS-ex- regarded as a psychostimulant action.
posed rats, and those were reversed by one-week treatment with CPE
(50, and 100 mg/kg, p.o.) or fluoxetine (10 mg/kg, p.o.) in the CUMS-
exposed rats. Two-way ANOVA followed by post-hoc Tukey’s multiple 3.4. Effect of CPE treatment in the CUMS rats on locomotion activity
comparison tests depicted significant differences in the number of
square crossing, and the number of rearing [F (5, 30) = 35.06, p < As shown in Fig. 3, the influences of CUMS and CPE on the loco-
0.0001] in the OFT. Rats in the CUMS group demonstrated a significant motion were also investigated using actophotometer. Application of
decline in the number of squares crossed (p < 0.0001; Fig. 2) and the two-way ANOVA depicted a significant difference in the locomotion
number of rearing (p < 0.01; Fig. 2) in the OFT as compared to the count [F (5, 25) = 18.58, p < 0.0001]. Post-hoc Tukey’s multiple
normal control rats. Treatment with CPE (100 mg/kg, p.o.) or fluox- comparison tests depicted that CUMS group showed a significant re-
etine (10 mg/kg, p.o.) for seven consecutive days significantly in- duction in the locomotion count as compared to the normal control rats
creased the number of squares crossed (p < 0.001, p < 0.0001; Fig. 2), in the actophotometer (p < 0.0001; Fig. 3). CPE (50, and 100 mg/kg,
and the number of rearing (p < 0.05, p < 0.05; Fig. 2) respectively in p.o.) or fluoxetine (10 mg/kg, p.o.) treatment for seven consecutive
the OFT as compared to the CUMS-exposed rats. CPE (50 mg/kg, p.o.) days significantly increased the locomotion count (p < 0.05, p <
also exerted significant reversal in the number of squares crossed (p < 0.001, p < 0.001; Fig. 3) in the actophotometer as compared to the
0.01; Fig. 2) in the OFT as compared to the CUMS-exposed rats. How- CUMS-exposed rats. However, no significant reversal in the locomotor
ever, no significant effects on the number of squares crossed, and the count was exerted after treatment with CPE (25 mg/kg, p.o.) as com-
number of rearing were observed on the treatment with a lower dose of pared with CUMS-exposed rats (p > 0.05; Fig. 3).
CPE (25 mg/kg, p.o.) when compared to the vehicle-treated CUMS-ex-
posed rats (p > 0.05; Fig. 2). Furthermore, there were no significant

1702
G.L. Gupta, J. Fernandes Biomedicine & Pharmacotherapy 109 (2019) 1698–1708

Fig. 4. Influences of CUMS and CPE on the


percent of sucrose preference. After the ad-
ministration of CPE (25, 50, and 100 mg/kg,
p.o.), fluoxetine (10 mg/kg, p.o.) or vehicle,
rats were subjected to the SPT. Results are
shown as mean ± SEM (n = 6) by vertical
bars, where ***p < 0.001 (Compared to
normal control group), #p < 0.05, ##p <
0.01, ###p < 0.001 (Compared to vehicle-
treated CUMS-exposed rats), Two-way analysis
of variance (ANOVA), followed by post-hoc
Tukey’s multiple comparison test. CUMS,
chronic unpredictable mild stress; CPE,
methanolic extract of areal part of Convolvulus
pluricaulis; SPT, sucrose preference test.

3.5. Influences of CPE on the percent of sucrose consumption differences in the pro-inflammatory cytokine IL-1β [F (5, 25) = 13.86,
p < 0.0001], IL-6 [F (5, 25) = 7.023, p < 0.0001], and TNFα [F (5,
As presented in Fig. 4, significant differences were depicted among 25) = 12.53, p < 0.0001] in the plasma of rats as compared to the
the treatments groups on the percent of sucrose consumption [F (5, normal control group. The post-hoc Tukey’s multiple comparison test
25) = 12.23, p < 0.0001]. Two-way ANOVA followed by post-hoc demonstrated that exposure of the rats to the chronic CUMS depicted
Tukey’s multiple comparison tests demonstrated the significant de- significant elevation in the pro-inflammatory cytokine IL-1β (p <
crease in the percent of sucrose consumption in the CUMS-exposed rats 0.0001; Fig. 6A), IL-6 (p < 0.001; Fig. 6B), and TNFα (p < 0.0001;
in comparison with normal control group (p < 0.001; Fig. 4). In this Fig. 6C) levels in the plasma. One-week treatment with CPE (50, and
study, one-week administration of CPE (50, and 100 mg/kg, p.o.) and 100 mg/kg, p.o.) and fluoxetine (10 mg/kg, p.o.) markedly attenuated
fluoxetine (10 mg/kg, p.o.) revealed an antidepressant-like effect, as IL-1β (p < 0.05, p < 0.0001, p < 0.001; Fig. 6A), IL-6 (p < 0.05, p <
percentage sucrose intake in CUMS-exposed rats significantly raised 0.01, p < 0.05; Fig. 6B), and TNFα (p < 0.01, p < 0.0001, p < 0.01;
(p < 0.05, p < 0.01, p < 0.001; Fig. 4) respectively as compared with Fig. 6C) levels in the plasma respectively as compared to the vehicle-
vehicle-treated CUMS-exposed rats. However, no significant alteration treated CUMS-exposed rats. Moreover, CPE (25 mg/kg, p.o.) failed to
was observed in the percentage sucrose consumption on the treatment produce any significant effects on the pro-inflammatory cytokine levels
with CPE (25 mg/kg, p.o.) as compared with the vehicle-treated CUMS- in the CUMS-exposed rats (p > 0.05; Fig. 6).
exposed rats (p > 0.05; Fig. 4).

3.6. Influences of CPE on depressive-like behavior in rats 3.8. Influences of CPE on the liver biomarkers

In the FST (Fig. 5), two-way ANOVA revealed significant differences The effect of CPE on the liver biomarkers were examined and pre-
among various groups in the immobility time [F (5, 25) = 49.52, p < sented in the Fig. 7. Two-way ANOVA revealed significant differences
0.0001]. Post-hoc Tukey’s multiple comparison tests demonstrated that in the liver biomarkers ALT [F (5, 25) = 6.11, p = 0.0008], AST [F (5,
CUMS-exposed rats depicted the significant increase in the immobility 25) = 11.71, p < 0.0001] and insignificant changes in the ALP levels
time as compared to the normal control rats (p < 0.0001; Fig. 5). [F (5, 25) = 1.47, p=0.2350] in the plasma of rats as compared to the
Chronic treatment with the CPE (50, and 100 mg/kg, p.o.) and fluox- normal control group. One-week CPE (100 mg/kg, p.o.) or fluoxetine
etine (10 mg/kg, p.o.) significantly decreased the immobility duration (10 mg/kg, p.o.) administration significantly decreased the elevated
in the CUMS-exposed rats when compared with the vehicle-treated level of liver enzymes ALT (p < 0.01, p < 0.05; Fig. 7A) and AST (p <
CUMS-exposed rats (p < 0.05, p < 0.0001, p < 0.0001; Fig. 5) re- 0.01, p < 0.05; Fig. 7B) levels respectively in the plasma as compared
spectively in the FST rat model. Moreover, CPE (25 mg/kg, p.o.) did not to the vehicle-treated CUMS-exposed rats. Moreover, CPE (50 mg/kg)
exert any significant differences in the duration of immobility in the showed significant changes on the ALT levels (p < 0.05; Fig. 7A) while
CUMS-exposed rats (p > 0.05; Fig. 5). AST and ALP were not changed significantly in the CUMS-exposed rats
(p > 0.05; Fig. 7B & 7C). However, CPE (25 mg/kg, p.o.) did not de-
3.7. Influences of CPE on the pro-inflammatory cytokines monstrate any significant changes on the ALT, AST, and ALP levels in
the CUMS-exposed rats (p > 0.05; Fig. 7). ALP levels were not changed
Next, the effect of CPE on the pro-inflammatory cytokine levels in significantly in the all animal group (p > 0.05; Fig. 7C).
the plasma were examined. Two-way ANOVA exerted significant

Fig. 5. Influences of CUMS and CPE on the


immobility time in the FST. After the adminis-
tration of CPE (25, 50, and 100 mg/kg, p.o.),
fluoxetine (10 mg/kg, p.o.) or vehicle, rats
were subjected to the FST. Results are shown as
mean ± SEM (n = 6) by vertical bars, where
****
p < 0.0001 (Compared to normal control
group), #p < 0.05, ####p < 0.0001
(Compared to vehicle-treated CUMS-exposed
rats), Two-way analysis of variance (ANOVA),
followed by post-hoc Tukey’s multiple com-
parison test. CUMS, chronic unpredictable mild
stress; CPE, methanolic extract of areal part of
Convolvulus pluricaulis; FST, forced swimming
test.

1703
G.L. Gupta, J. Fernandes Biomedicine & Pharmacotherapy 109 (2019) 1698–1708

comparison test exerted that CUMS-exposed rats demonstrated sig-


nificant decline in the noradrenaline levels in the hippocampus (p <
0.0001; Fig. 9A), and prefrontal cortex (p < 0.0001; Fig. 9B) as com-
pared to the normal control group. One-week CPE (50, and 100 mg/kg,
p.o.) or fluoxetine (10 mg/kg, p.o.) administration reversed the level of
noradrenaline in the hippocampus (p < 0.05, p < 0.0001, p < 0.001;
Fig. 9A) as well as in the prefrontal cortex (p < 0.05, p < 0.0001, p <
0.05; Fig. 9B) respectively in the CUMS-exposed rats as compared to the
vehicle-treated CUMS-exposed rats, therefore exerts the antidepressant-
like effects.

4. Discussion

In the current study, we assessed the antidepressant-like effect of


CPE and also investigated the possible mechanism using a CUMS ex-
posure rat model of depression. Our major findings reveal that CUMS
resulted in increased depression-like behavior, inflammation, liver
biomarkers and decreased serotonin, noradrenaline level in the hippo-
campus as well as in the prefrontal cortex of the rats. CPE-treatment
efficiently ameliorated CUMS-induced depression, pro-inflammatory
cytokines, liver biomarkers, and brain monoaminergic neuro-
transmitters.
The CUMS model is an acclaimed and authentic animal model of
depression with high translational potential because this model can
mimic the core symptoms of the depression by exposing animals to
diverse kind of stress every day [52,63]. Hence, we selected it to re-
search the antidepressant-like effects of CPE in this study. However, the
influence of CPE on depression-like behaviors have been investigated
earlier in other animal models [37].
Moreover, our results revealed that rats exposed to CUMS for 27
consecutive days, demonstrated a remarkable decrease in the sucrose
Fig. 6. Influences of CUMS and CPE on pro-inflammatory cytokines in the blood
intake in the SPT, immobility time in the FST, a number of line cross-
plasma. (6 A) IL-1β levels, (6B) IL-6 levels (6C) TNF-α levels. Results are shown
ings, rearing in the OFT, locomotor index in the actophotometer, and
as mean ± SEM (n = 6) by vertical bars, where ***p < 0.001, ****p <
0.0001 (Compared to normal control group), #p < 0.05, ##p < 0.01
regarded as induction of depressive-like behavior in rats. In the earlier
###
p < 0.001, ####p < 0.0001 (Compared to vehicle-treated CUMS-ex- reports, the SPT is used to demonstrate the anhedonia-like behavior in
posed rats), Two-way analysis of variance (ANOVA), followed by post-hoc animals (a decrease of interest to rewards) and, considered as a core
Tukey’s multiple comparison test. CUMS, chronic unpredictable mild stress; symptom of depression [47,52,64]. In the SPT, rats were given access to
CPE, methanolic extract of areal part of Convolvulus pluricaulis; IL-1β, inter- a highly preferred sucrose solution, or to a choice between a sucrose
leukin-1β; IL-6, interleukin-6; TNF-α, tumor necrosis factor-α. solution and drinking water. Sucrose preference decreases over weeks
of CUMS exposure but can be restored to normal levels by chronic
3.9. Influences of CPE on the serotonin levels in the hippocampus and treatment with antidepressant drugs [5,15]. Furthermore, we found
prefrontal cortex that one-week consecutive CPE (50 and 100 mg/kg) or fluoxetine
(10 mg/kg) treatment significantly increased sucrose preference index
As shown in Fig. 8, two-way ANOVA depicted significant differences and effectively attenuated the depressive-like behavior of CUMS-ex-
among the treatments groups on the serotonin levels in the hippo- posed rats.
campus [F (5, 25) = 49.69, p < 0.0001], and prefrontal cortex [F (5, The FST was used to evaluate the antidepressant-like effect of the
25) = 36.96, p < 0.0001]. Post-hoc Tukey’s multiple comparison test drugs because it is a reliable animal model with high predictive validity
showed that CUMS-exposed rats exhibited significant decrement in the [59–61]. In the present study, we observed an increment of the im-
serotonin levels in the hippocampus (p < 0.0001; Fig. 8A), and pre- mobility time in the FST in the CUMS-exposed rats. It is well reported
frontal cortex (p < 0.0001; Fig. 8B) as compared to the normal control earlier that rodents exposed to CUMS display despair behavior and our
group. One-week CPE (50, and 100 mg/kg, p.o.) or fluoxetine (10 mg/ results are also consistent with the previous studies [15,65]. One-week
kg, p.o.) administration improved the level of serotonin in the hippo- CPE (50 and 100 mg/kg) or fluoxetine (10 mg/kg) treatment sig-
campus (p < 0.01, p < 0.0001, p < 0.0001; Fig. 8A) as well as in the nificantly declined immobility time in the FST, indicating anti-
prefrontal cortex (p < 0.01, p < 0.0001, p < 0.0001; Fig. 8B) re- depressant-like effects of CPE in the CUMS animal model, whereas
spectively in the CUMS-exposed rats as compared to the vehicle-treated 25 mg/kg of CPE did not alter the CUMS-induced increase in the im-
CUMS-exposed rats, therefore demonstrates the antidepressant-like mobility time in the FST. Our results are in concordance with earlier
activity. reports which reveal that CPE exerts an antidepressant effect in other
animal models of depression [37]. Therefore, the results confirmed the
antidepressant-like activity of CPE in the CUMS animal model.
3.10. Influences of CPE on the noradrenaline levels in the hippocampus and We, further, confirmed that whether the decreased immobility in
prefrontal cortex the FST on the administration of CPE was presented by the anti-
depressant-like effect or the locomotor-stimulant activity. Hence we
As presented in Fig. 9, two-way ANOVA revealed significant dif- used the OFT and actophotometer animal models to evaluate the lo-
ferences among the treatments groups on the noradrenaline levels in comotor, exploratory behaviors [53,54]. In this study, CUMS-exposed
the hippocampus [F (5, 25) = 55.95, p < 0.0001], and prefrontal rats presented significant impairment in the number of crossing, rearing
cortex [F (5, 25) = 22.77, p < 0.0001]. Post-hoc Tukey’s multiple in the OFT and locomotor count in the actophotometer. Treating CUMS-

1704
G.L. Gupta, J. Fernandes Biomedicine & Pharmacotherapy 109 (2019) 1698–1708

Fig. 7. Influences of CUMS and CPE on liver


biomarkers in the blood plasma. (7 A) ALT le-
vels, (7B) AST levels (7C) ALP levels. Results
are shown as mean ± SEM (n = 6) by vertical
bars, where **p < 0.01, ***p < 0.001
(Compared to normal control group), #p <
0.05, ##p < 0.01 (Compared to vehicle-
treated CUMS-exposed rats), Two-way analysis
of variance (ANOVA), followed by post-hoc
Tukey’s multiple comparison test. CUMS,
chronic unpredictable mild stress; CPE, me-
thanolic extract of areal part of Convolvulus
pluricaulis; ALT, Alanine transaminase; AST,
Aspartate transaminase, and ALP, Alkaline
phosphatase.

group, suggesting that CPE administration have no influences on the


locomotor activity in rats. These results are in accordance with the
previous studies implying that the antidepressant-like effect of CPE may
be uninvolved by the locomotor-stimulant activity [66].
Interestingly, previous studies have indicated that chronic stress
plays a crucial role in the progression of depression, associated in-
flammation, and disturbance of brain neurotransmitters [20,63,67,68].
Numerous investigations have confirmed that increased inflammatory
cytokines in the brain played a crucial role in the pathogenesis of de-
pression [20,63,69–72]. Therefore, pro-inflammatory cytokines might
be the apparent target for the treatment of depression. Previous studies
have reported that the cytokine-induced behavioral alterations are as-
sociated with changes in the metabolism of serotonin levels in the
hippocampus and prefrontal cortex [4,20,26,71,73,74]. Therefore, we
thought to measure the levels of cytokines and serotonin in the CUMS-
exposed rats, and following treatment with CPE and fluoxetine. Our
results revealed that CUMS markedly aggravated pro-inflammatory
markers IL-1β, IL-6, TNF-α, and depression-like behavior. These find-
ings are consistent with those analogous results obtained after similar
paradigms of CUMS [75]. Clinical studies also revealed that pro-in-
flammatory cytokines, including IL-1β, IL-6, and TNF-α were sig-
Fig. 8. Effects of CUMS and CPE on the serotonin level in the (8 A) hippo- nificantly elevated during the depression [76–78]. To further elucidate
campus, and (8B) prefrontal cortex in the CUMS-exposed rats. Results are the pharmacological mechanism of CPE, the current investigation
shown as mean ± SEM (n = 6) by vertical bars, where ****p < 0.0001 probed that one-week treatment of CPE could reverse the inflammatory
(Compared to normal control group), ##p < 0.01, ####p < 0.0001 cytokines levels induced by the CUMS exposure. These results revealed
(Compared to vehicle-treated CUMS-exposed rats), Two-way analysis of var-
that CPE (50 and 100 mg/kg) or fluoxetine (10 mg/kg) exerted an anti-
iance (ANOVA), followed by post-hoc Tukey’s multiple comparison test. CUMS,
inflammatory and neuroprotective effect. Interestingly, CPE treatment
chronic unpredictable mild stress; CPE, methanolic extract of areal part of
Convolvulus pluricaulis. at 100 mg/kg completely reversed the CUMS-induced increase in the IL-
1β, IL-6, and TNF-α levels as compared with CUMS-exposed rats. This is
the first report for the action of CPE to ameliorate the pro-inflammatory
exposed rats with CPE (50 and 100 mg/kg) or fluoxetine (10 mg/kg) cytokines, including IL-1β, IL-6, and TNF-α levels in the CUMS asso-
restored their locomotion, as evident from the significantly higher ciated inflammatory diseases. However, the effects of CPE on the
number of line crossings, rearing in the OFT and remarkable restoration CUMS-induced depression-like behavior and altered cytokines have
of the locomotion count in the actophotometer. However, in normal been unscreened yet. We speculated that CPE also inhibited in-
control rats, one-week treatment with all doses of CPE did not alter the flammation in the CUMS rats via modulating pro-inflammatory
locomotor activity compared to the vehicle-treated normal control

1705
G.L. Gupta, J. Fernandes Biomedicine & Pharmacotherapy 109 (2019) 1698–1708

of the standard drug fluoxetine.


On other way, studies on depression are mainly engrossed on the
changes in the brain, but not on the liver. On a primary level, we also
measured whether CUMS alter the liver enzymes and also screening the
influences of CPE in the altered liver biomarkers. In our study, one-
week CPE (100 mg/kg) or fluoxetine (10 mg/kg) treatments sig-
nificantly declined the increased levels of liver enzymes ALT and AST
induced by CUMS, indicating CPE was capable of ameliorating the
hepatic injury. Our finding also supports the previous reports on the
elevation of liver enzymes during CUMS [5,82]. It was also suggested
that depression could be linked with liver injury in the earlier reports
[5,83,84], and therefore, CPE can also ameliorate depressive-like be-
havior by inhibiting liver-brain inflammation axis.

5. Conclusions

Based on our results, we conclude that CUMS exposure instigated


depression-like behavior as well as alteration in inflammatory cyto-
kines, liver enzymes, serotonin and noradrenaline levels.
Administration of CPE attenuated CUMS-induced depressive-like be-
havior. The antidepressant-like effect of CPE in the CUMS-exposed rats
may be linked to the alteration of inflammatory cytokines, liver en-
zymes, serotonin and noradrenaline levels. CPE may be a potential drug
for the treatment of depression and its associated neuroinflammation.
However, further studies are needed to explore the gene and protein
expression alteration to understand the molecular mechanism of CPE
effects in CUMS-induced depression.

Conflict of interest

The authors declare no conflict of interest.

Fig. 9. Effects of CUMS and CPE on the noradrenaline level in the (9 A) hip-
Funding information
pocampus, and (9B) prefrontal cortex in the CUMS-exposed rats. Results are
shown as mean ± SEM (n = 6) by vertical bars, where ****p < 0.0001
(Compared to normal control group), #p < 0.05, ###p < 0.001, ####p <
Authors are grateful to SVKM’s NMIMS (Deemed to be University),
0.0001 (Compared to vehicle-treated CUMS-exposed rats), Two-way analysis of Mumbai, India for providing a grant for the research.
variance (ANOVA), followed by post-hoc Tukey’s multiple comparison test.
CUMS, chronic unpredictable mild stress; CPE, methanolic extract of areal part Appendix A. Supplementary data
of Convolvulus pluricaulis.
Supplementary material related to this article can be found, in the
cytokines associated with serotonin and noradrenaline decline. IL-1β, online version, at doi:https://doi.org/10.1016/j.biopha.2018.11.046.
IL-6, and TNF-α are the most widely investigated cytokines as a potent
mediator of inflammation and associated depression. The involvement References
of IL-1β, IL-6, and TNF-α cytokines to ingress the brain via leaky re-
[1] X.L. Zhu, J.J. Chen, F. Han, C. Pan, T.T. Zhuang, Y.F. Cai, Y.P. Lu, Novel anti-
gions in the blood-brain barrier has been demonstrated earlier in the
depressant effects of Paeonol alleviate neuronal injury with concomitant alterations
pathogenesis of the depression [20,70,71]. These cytokines also dys- in BDNF, Rac1 and RhoA levels in chronic unpredictable mild stress rats,
regulated the metabolism of relevant neurotransmitters like serotonin, Psychopharmacology (Berl.) (2018) 1–15, https://doi.org/10.1007/s00213-018-
noradrenaline, and disruption of synaptic plasticity through alterations 4915-7.
[2] World Health Organization, Depression and other common mental disorders: global
brain-derived neurotrophic factor. However, activation of the kynur- health estimates, World Heal. Organ. (2017) 1–24 doi:CC BY-NC-SA 3.0 IGO.
enine pathway of tryptophan metabolism may be involved in the pa- [3] N. Eshel, J.P. Roiser, Reward and punishment processing in depression, Biol.
thogenesis of the depression (Bryleva and Brundin, 2016; O'Connor Psychiatry 68 (2010) 118–124, https://doi.org/10.1016/j.biopsych.2010.01.027.
[4] Z.Q. Li, Z.Y. Yan, F.J. Lan, Y.Q. Dong, Y. Xiong, Suppression of NLRP3 inflamma-
et al., 2009). some attenuates stress-induced depression-like behavior in NLGN3-deficient mice,
Increasing evidence suggested that monoaminergic deficiency has Biochem. Biophys. Res. Commun. 501 (2018) 933–940, https://doi.org/10.1016/j.
been regarded to underlie depressive disorders and most of the existing bbrc.2018.05.085.
[5] H.M. Jia, Q. Li, C. Zhou, M. Yu, Y. Yang, H.W. Zhang, G. Ding, H. Shang, Z.M. Zou,
antidepressants work by increasing the serotonin and/or noradrenaline Chronic unpredictive mild stress leads to altered hepatic metabolic profile and gene
levels [79–81]. In the present study, we also observed a significant expression, Sci. Rep. 6 (2016) 1–10, https://doi.org/10.1038/srep23441.
decline of serotonin and noradrenaline levels in the hippocampus as [6] D.M. Kokare, P.S. Singru, M.P. Dandekar, C.T. Chopde, N.K. Subhedar, Involvement
of alpha-melanocyte stimulating hormone (α-MSH) in differential ethanol exposure
well as in the prefrontal cortex of the CUMS-exposed rats. Further, one and withdrawal related depression in rat: neuroanatomical-behavioral correlates,
week CPE (50 and 100 mg/kg) or fluoxetine (10 mg/kg) treatment Brain Res. 1216 (2008) 53–67, https://doi.org/10.1016/j.brainres.2008.03.064.
significantly reversed the serotonin and noradrenaline levels in the [7] V. Krishnan, E.J. Nestler, The molecular neurobiology of depression, Nature 455
(2008) 894–902, https://doi.org/10.1038/nature07455.
hippocampus as well as in the prefrontal cortex of the CUMS-exposed
[8] K. Yan, Y.-B. Chen, J.-R. Wu, K.-D. Li, Y.-L. Cui, Current rapid-onset antidepressants
rats, stipulating that the antidepressant-like effect of CPE could be due and related animal models, Curr. Pharm. Des. 24 (2018), https://doi.org/10.2174/
to the improvement of major monoamine neurotransmitters serotonin 1381612824666180727115222.
and noradrenaline, these results are also consistent with the previous [9] D.M. Gerhard, E.S. Wohleb, R.S. Duman, Emerging treatment mechanisms for de-
pression: focus on glutamate and synaptic plasticity, Drug Discov. Today 21 (2016)
reports [37]. The results of CPE were found to be comparable with that 454–464, https://doi.org/10.1016/j.drudis.2016.01.016.

1706
G.L. Gupta, J. Fernandes Biomedicine & Pharmacotherapy 109 (2019) 1698–1708

[10] D.M. Gerhard, R.S. Duman, Rapid-acting antidepressants: mechanistic insights and [33] P. Rachitha, K. Krupashree, G.V. Jayashree, H.K. Kandikattu, N. Amruta,
future directions, Curr. Behav. Neurosci. Reports. 5 (2018) 36–47, https://doi.org/ N. Gopalan, M.K. Rao, F. Khanum, Chemical composition, antioxidant potential,
10.1016/j.chemosphere.2012.12.037.Reactivity. macromolecule damage and neuroprotective activity of Convolvulus pluricaulis, J.
[11] J.P. Pandarakalam, Challenges of treatment-resistant depression, Psychiatr. Danub. Tradit. Complement. Med. 8 (2018) 483–496, https://doi.org/10.1016/j.jtcme.
30 (2018) 273–284, https://doi.org/10.24869/psyd.2018.273. 2017.11.002.
[12] R. Chockalingam, B.M. Gott, C.R. Conway, Tricyclic Antidepressants and [34] J. Malik, M. Karan, K. Vasisht, Attenuating effect of bioactive coumarins from
Monoamine Oxidase Inhibitors: Are They Too Old for a New Look? (2018), https:// Convolvulus pluricaulis on scopolamine-induced amnesia in mice, Nat. Prod. Res.
doi.org/10.1007/164_2018_133. 30 (2016) 578–582, https://doi.org/10.1080/14786419.2015.1025398.
[13] T.C. Lee, A.M. Alessio, R.M. Miyaoka, P.E. Kinahan, Morphology supporting func- [35] J. Malik, S. Choudhary, P. Kumar, Protective effect of Convolvulus pluricaulis
tion: Attenuation correction for SPECT/CT, PET/CT, and PET/MR imaging, Q. J. standardized extract and its fractions against 3-nitropropionic acid-induced neu-
Nucl. Med. Mol. Imaging 60 (2016) 25–39, https://doi.org/10.1016/j. rotoxicity in rats, Pharm. Biol. 53 (2015) 1448–1457, https://doi.org/10.3109/
chemosphere.2012.12.037.Reactivity. 13880209.2014.984856.
[14] J.W. Murrough, C.G. Abdallah, S.J. Mathew, Targeting glutamate signalling in [36] M. Heba, S. Faraz, S. Banerjee, Effect of Shankhpushpi on alcohol addiction in mice,
depression: progress and prospects, Nat. Rev. Drug Discov. 16 (2017) 472–486, Pharmacogn. Mag. 13 (2017) S148–S153, https://doi.org/10.4103/0973-1296.
https://doi.org/10.1038/nrd.2017.16. 203976.
[15] T. Qin, F. Fang, M. Song, R. Li, Z. Ma, S. Ma, Umbelliferone reverses depression-like [37] D. Dhingra, R. Valecha, Evaluation of the antidepressant-like activity of
behavior in chronic unpredictable mild stress-induced rats by attenuating neuronal Convolvulus pluricaulis choisy in the mouse forced swim and tail suspension tests,
apoptosis via regulating ROCK/Akt pathway, Behav. Brain Res. 317 (2017) Med. Sci. Monit. 13 (2007) BR155–61 http://www.ncbi.nlm.nih.gov/pubmed/
147–156, https://doi.org/10.1016/j.bbr.2016.09.039. 17599020.
[16] M. Berk, L.J. Williams, F.N. Jacka, A.O. Neil, J.A. Pasco, S. Moylan, N.B. Allen, [38] G. Garg, A. Patil, J. Singh, N. Kaushik, A. Praksah, A. Pal, A. Chakrabarti,
A.L. Stuart, A.C. Hayley, M.L. Byrne, M. Maes, So depression is an inflammatory Pharmacological evaluation of Convolvulus pluricaulis as hypolipidaemic agent in
disease, but where does the inflammation come from? BMC Med. 11 (2013) 200, Triton WR-1339-induced hyperlipidaemia in rats, J. Pharm. Pharmacol. 70 (2018)
https://doi.org/10.1186/1741-7015-11-200. 1572–1580, https://doi.org/10.1111/jphp.13004.
[17] S.Q. Dong, Q.P. Zhang, J.X. Zhu, M. Chen, C.F. Li, Q. Liu, D. Geng, L.T. Yi, [39] Sristi Verma, Reema Sinha, Puspendra Kumar, Faizal Amin, Jainendra Jain,
Gypenosides reverses depressive behavior via inhibiting hippocampal neuroin- Shivani Tanwar, Study of Convolvulus pluricaulis for antioxidant and antic-
flammation, Biomed. Pharmacother. 106 (2018) 1153–1160, https://doi.org/10. onvulsant activity, Cent. Nerv. Syst. Agents Med. Chem. 12 (2012) 55–59, https://
1016/j.biopha.2018.07.040. doi.org/10.2174/187152412800229161.
[18] H. Jiang, G. Zheng, J. Lv, H. Chen, J. Lin, Y. Li, G. Fan, X. Ding, Identification of [40] A. Dhar, S.K. Maurya, A. Mishra, G.K. Singh, M.K. Singh, A. Seth, Preliminary
Centella asiatica’s effective ingredients for inducing the neuronal differentiation, screening of a classical ayurvedic formulation for anticonvulsant activity, Anc. Sci.
evidence-based complement, Altern. Med. 2016 (2016), https://doi.org/10.1155/ Life 36 (2016) 28–34, https://doi.org/10.4103/0257-7941.195410.
2016/9634750. [41] A. Nahata, U.K. Patil, V.K. Dixit, Anxiolytic activity of Evolvulus alsinoides and
[19] C. Fan, Q. Song, P. Wang, Y. Li, M. Yang, B. Liu, S.Y. Yu, Curcumin protects against Convulvulus pluricaulis in rodents, Pharm. Biol. 47 (2009) 444–451, https://doi.org/
chronic stress-induced dysregulation of neuroplasticity and depression-like beha- 10.1080/13880200902822596.
viors via suppressing IL-1β pathway in rats, Neuroscience (2018), https://doi.org/ [42] K. Sairam, C.V. Rao, R.K. Goel, Effect of Convolvulus pluricaulis Chois on gastric
10.1016/j.neuroscience.2018.09.028. ulceration and secretion in rats, Indian J. Exp. Biol. 39 (2001) 350–354 (accessed
[20] C.L. Raison, L. Capuron, A.H. Miller, Cytokines sing the blues: inflammation and the September 20, 2018), http://www.ncbi.nlm.nih.gov/pubmed/11491580.
pathogenesis of depression, Trends Immunol. 27 (2006) 24–31, https://doi.org/10. [43] N.K. Sethiya, S. Mishra, Review on ethnomedicinal uses and phyto-pharmacology of
1016/j.it.2005.11.006. memory boosting herb Convolvulus pluricaulis Choisy, Aust. J. Med. Herbal. 22
[21] Y.M. Liu, J.D. Shen, L.P. Xu, H.B. Li, Y.C. Li, L.T. Yi, Ferulic acid inhibits neuro- (2010) 19–25 http://www.encognitive.com/files/Reviewonethnomedicinalusesand
inflammation in mice exposed to chronic unpredictable mild stress, Int. phytopharmacologyofmemoryboostingherbConvolvuluspluricaulisChoisy.pdf.
Immunopharmacol. 45 (2017) 128–134, https://doi.org/10.1016/j.intimp.2017. [44] P. Agarwal, B. Sharma, A. Fatima, S.K. Jain, An update on Ayurvedic Herb
02.007. Convolvulus pluricaulis Choisy, Asian Pac. J. Trop. Biomed. 4 (2014) 245–252,
[22] J.C. Felger, Z. Li, E. Haroon, B.J. Woolwine, M.Y. Jung, X. Hu, A.H. Miller, https://doi.org/10.1016/S2221-1691(14)60240-9.
Inflammation is associated with decreased functional connectivity within corti- [45] J.C. Capra, M.P. Cunha, D.G. Machado, A.D.E. Zomkowski, B.G. Mendes,
costriatal reward circuitry in depression, Mol. Psychiatry 21 (2016) 1358–1365, A.R.S. Santos, M.G. Pizzolatti, A.L.S. Rodrigues, Antidepressant-like effect of sco-
https://doi.org/10.1038/mp.2015.168. poletin, a coumarin isolated from Polygala sabulosa (Polygalaceae) in mice: evi-
[23] C. Yang, F.J. Bosker, J. Li, R.A. Schoevers, N-acetylcysteine as add-on to anti- dence for the involvement of monoaminergic systems, Eur. J. Pharmacol. 643
depressant medication in therapy refractory major depressive disorder patients with (2010) 232–238, https://doi.org/10.1016/j.ejphar.2010.06.043.
increased inflammatory activity: Study protocol of a double-blind randomized [46] H.-J. Kim, S. Il Jang, Y.-J. Kim, H.-T. Chung, Y.-G. Yun, T.-H. Kang, O.-S. Jeong, Y.-
placebo-controlled trial, BMC Psychiatry 18 (2018) 279, https://doi.org/10.1186/ C. Kim, Scopoletin suppresses pro-inflammatory cytokines and PGE2 from LPS-sti-
s12888-018-1845-1. mulated cell line, RAW 264.7 cells, Fitoterapia 75 (2004) 261–266, https://doi.org/
[24] F.P. MacMaster, N. Carrey, L.M. Langevin, N. Jaworska, S. Crawford, Disorder- 10.1016/j.fitote.2003.12.021.
specific volumetric brain difference in adolescent major depressive disorder and [47] P. Willner, The chronic mild stress (CMS) model of depression: history, evaluation
bipolar depression, Brain Imaging Behav. 8 (2014) 119–127, https://doi.org/10. and usage, Neurobiol. Stress 6 (2017) 78–93, https://doi.org/10.1016/j.ynstr.2016.
1007/s11682-013-9264-x. 08.002.
[25] W.-Y. Zhang, K.-Y. Wang, Y.-J. Li, Y.-R. Li, R.-Z. Lu, Chronic stress causes protein [48] W.C. Evans, D. Evans, G.E. Trease, Trease and Evans Pharmacognosy, Saunders/
kinase C epsilon-aldehyde dehydrogenase 2 signaling pathway perturbation in the Elsevier, 2009.
rat hippocampus and prefrontal cortex, but not in the myocardium, Neural Regen. [49] H. Amin, R. Sharma, M. Vyas, P.K. Prajapati, K. Dhiman, Shankhapushpi
Res. 13 (2018) 1225–1230, https://doi.org/10.4103/1673-5374.235060. (Convolvulus pluricaulis Choisy): validation of the Ayurvedic therapeutic claims
[26] Y. Zhang, K. Fan, Y. Liu, G. Liu, X. Yang, J. Ma, Cathepsin C aggravates neuroin- through contemporary studies, Int. J. Green Pharm. (2014) 193–200, https://doi.
flammation involved in disturbances of behaviour and neurochemistry in acute and org/10.4103/0973-8258.142666.
chronic stress-induced murine model of depression, Neurochem. Res. 43 (2018) [50] OECD, Guideline 423. Acute Oral Toxicity- Acute Toxic Class Method. 470 Adopted
1702, https://doi.org/10.1007/s11064-018-2564-1. by the Council on 17th, December 2001, 2001, Organization for economic co-
[27] K. Sharma, M. Bhatnagar, S.K. Kulkarni, Effect of Convolvulus pluricaulis Choisy operation and development (OECD), 2001 (accessed October 1, 2018), https://ntp.
and Asparagus racemosus Willd on learning and memory in young and old mice: a niehs.nih.gov/iccvam/suppdocs/feddocs/oecd/oecd_gl423.pdf.
comparative evaluation, Indian J. Exp. Biol. 48 (2010) 479–485 (Accessed 19 [51] D.G. MacHado, M.P. Cunha, V.B. Neis, G.O. Balen, A. Colla, J. Grando,
October 2018), http://www.ncbi.nlm.nih.gov/pubmed/20795365. P.S. Brocardo, L.E.B. Bettio, J.C. Capra, A.L.S. Rodrigues, Fluoxetine reverses de-
[28] H. Amin, R. Sharma, P. Prajapati, R. Dwivedi, H. Vyas, M. Vyas, Nootropic pressive-like behaviors and increases hippocampal acetylcholinesterase activity
(medhya) effect of Bhāvita Śaṇkhapuṣpī tablets: a clinical appraisal, Anc. Sci. Life induced by olfactory bulbectomy, Pharmacol. Biochem. Behav. 103 (2012)
34 (2014) 109–112, https://doi.org/10.4103/0257-7941.153476. 220–229, https://doi.org/10.1016/j.pbb.2012.08.024.
[29] P.A. Kizhakke, S. Olakkaran, A. Antony, K.S. Tilagul, P.G. Hunasanahally, [52] G.L. Wang, Y.P. Wang, J.Y. Zheng, L.X. Zhang, Monoaminergic and aminoacidergic
Convolvulus pluricaulis (Shankhapushpi) ameliorates human microtubule-asso- receptors are involved in the antidepressant-like effect of ginsenoside Rb1 in mouse
ciated protein tau (hMAPτ) induced neurotoxicity in Alzheimer’s disease Drosophila hippocampus (CA3) and prefrontal cortex, Brain Res. 1699 (2018) 44–53, https://
model, J. Chem. Neuroanat. (2017), https://doi.org/10.1016/j.jchemneu.2017.10. doi.org/10.1016/j.brainres.2018.05.035.
002. [53] X. Zu, M. Zhang, W. Li, H. Xie, Z. Lin, N. Yang, X. Liu, W. Zhang, Antidepressant-like
[30] S. Bihaqi, A. Singh, M. Tiwari, Supplementation of Convolvulus pluricaulis at- effect of Bacopaside I in mice exposed to chronic unpredictable mild stress by
tenuates scopolamine-induced increased tau and Amyloid precursor protein (AβPP) modulating the hypothalamic–pituitary–adrenal axis function and activating BDNF
expression in rat brain, Indian J. Pharmacol. 44 (2012) 593–598, https://doi.org/ signaling pathway, Neurochem. Res. 42 (2017) 3233–3244, https://doi.org/10.
10.4103/0253-7613.100383. 1007/s11064-017-2360-3.
[31] S.W. Bihaqi, M. Sharma, A.P. Singh, M. Tiwari, Neuroprotective role of Convolvulus [54] L. Cai, R. Li, W.J. Tang, G. Meng, X.Y. Hu, T.N. Wu, Antidepressant-like effect of
pluricaulis on aluminium induced neurotoxicity in rat brain, J. Ethnopharmacol. geniposide on chronic unpredictable mild stress-induced depressive rats by reg-
124 (2009) 409–415, https://doi.org/10.1016/j.jep.2009.05.038. ulating the hypothalamus-pituitary-adrenal axis, Eur. Neuropsychopharmacol. 25
[32] M. Kaur, A. Prakash, A.N. Kalia, Neuroprotective potential of antioxidant potent (2015) 1332–1341, https://doi.org/10.1016/j.euroneuro.2015.04.009.
fractions from Convolvulus pluricaulis Chois. In 3-nitropropionic acid challenged [55] J.F. Ge, W.C. Gao, W.M. Cheng, W.L. Lu, J. Tang, L. Peng, N. Li, F.H. Chen, Orcinol
rats, Nutr. Neurosci. 19 (2016) 70–78, https://doi.org/10.1179/1476830515Y. glucoside produces antidepressant effects by blocking the behavioural and neuronal
0000000022. deficits caused by chronic stress, Eur. Neuropsychopharmacol. 24 (2014) 172–180,

1707
G.L. Gupta, J. Fernandes Biomedicine & Pharmacotherapy 109 (2019) 1698–1708

https://doi.org/10.1016/j.euroneuro.2013.05.007. [71] J.D. Rosenblat, J.M. Gregory, R.S. McIntyre, Pharmacologic implications of in-
[56] W. Song, Y. Guo, S. Jiang, L. Wei, Z. Liu, X. Wang, Y. Su, Antidepressant effects of flammatory comorbidity in bipolar disorder, Curr. Opin. Pharmacol. 29 (2016)
the Ginsenoside metabolite compound K, assessed by behavioral despair test and 63–69, https://doi.org/10.1016/j.coph.2016.06.007.
chronic unpredictable mild stress model, Neurochem. Res. 43 (2018) 1371–1382, [72] R. Haapakoski, J. Mathieu, K.P. Ebmeier, H. Alenius, M. Kivimäki, Cumulative
https://doi.org/10.1007/s11064-018-2552-5. meta-analysis of interleukins 6 and 1β, tumour necrosis factor α and C-reactive
[57] A. Kumar, A. Prakash, D. Pahwa, Galantamine potentiates the protective effect of protein in patients with major depressive disorder, Brain Behav. Immun. 49 (2015)
rofecoxib and caffeic acid against intrahippocampal Kainic acid-induced cognitive 206–215, https://doi.org/10.1016/j.bbi.2015.06.001.
dysfunction in rat, Brain Res. Bull. 85 (2011) 158–168, https://doi.org/10.1016/j. [73] E.A. Rahim, Z. Mohd-Zain, J. Hussaini, E.H. Wong, L.L. Nyein, N. Parasakthi,
brainresbull.2011.03.010. A. Adnan, N.K. Palanisamy, Adherence of Streptococcus pneumoniae and expres-
[58] M. Erburu, I. Muñoz-Cobo, T. Diaz-Perdigon, P. Mellini, T. Suzuki, E. Puerta, sion analysis of neuraminidase gene (NanA and NanB) after interaction of A549
R.M. Tordera, SIRT2 inhibition modulate glutamate and serotonin systems in the human lung epithelial cells with pneumococcal strains of various serotypes, Malays.
prefrontal cortex and induces antidepressant-like action, Neuropharmacology 117 J. Microbiol. 13 (2017) 210–216, https://doi.org/10.1007/978-0-585-37970-8_8.
(2017) 195–208, https://doi.org/10.1016/j.neuropharm.2017.01.033. [74] B. Sperner-Unterweger, C. Kohl, D. Fuchs, Immune changes and neurotransmitters:
[59] D.M. Kokare, M.P. Dandekar, P.S. Singru, G.L. Gupta, N.K. Subhedar, Involvement Possible interactions in depression? Prog. Neuro-Psychopharmacol. Biol.
of α-MSH in the social isolation induced anxiety- and depression-like behaviors in Psychiatry. 48 (2014) 268–276, https://doi.org/10.1016/j.pnpbp.2012.10.006.
rat, Neuropharmacology 58 (2010) 1009–1018, https://doi.org/10.1016/j. [75] X. Liu, C. Liu, Baicalin ameliorates chronic unpredictable mild stress-induced de-
neuropharm.2010.01.006. pressive behavior: involving the inhibition of NLRP3 inflammasome activation in
[60] R. Yawalkar, H. Changotra, G.L. Gupta, Protective influences of N-acetylcysteine rat prefrontal cortex, Int. Immunopharmacol. 48 (2017) 30–34, https://doi.org/10.
against alcohol abstinence-induced depression by regulating biochemical and 1016/j.intimp.2017.04.019.
GRIN2A, GRIN2B gene expression of NMDA receptor signaling pathway in rats, [76] P. Martinez, L. Lien, S. Zemore, J.G. Bramness, S.P. Neupane, Circulating cytokine
Neurochem. Int. 118 (2018) 73–81, https://doi.org/10.1016/j.neuint.2018.04.011. levels are associated with symptoms of depression and anxiety among people with
[61] R.D. Porsolt, G. Anton, N. Blavet, M. Jalfre, Behavioural despair in rats: a new alcohol and drug use disorders, J. Neuroimmunol. 318 (2018) 80–86, https://doi.
model sensitive to antidepressant treatments, Eur. J. Pharmacol. 47 (1978) org/10.1016/j.jneuroim.2018.02.011.
379–391, https://doi.org/10.1016/0014-2999(78)90118-8. [77] A. Dey, P. Hankey Giblin, Insights into macrophage heterogeneity and cytokine-
[62] J. Thomas, R. Khanam, D. Vohora, A validated HPLC-UV method and optimization induced neuroinflammation in major depressive disorder, Pharmaceuticals 11
of sample preparation technique for norepinephrine and serotonin in mouse brain, (2018) 64, https://doi.org/10.3390/ph11030064.
Pharm. Biol. 53 (2015) 1539–1544, https://doi.org/10.3109/13880209.2014. [78] C.A. Köhler, T.H. Freitas, B. Stubbs, M. Maes, M. Solmi, N. Veronese, N.Q. de
991837. Andrade, G. Morris, B.S. Fernandes, A.R. Brunoni, N. Herrmann, C.L. Raison,
[63] C. Zhang, Y.P. Zhang, Y.Y. Li, B.P. Liu, H.Y. Wang, K.W. Li, S. Zhao, C. Song, B.J. Miller, K.L. Lanctôt, A.F. Carvalho, Peripheral alterations in cytokine and
Minocycline ameliorates depressive behaviors and neuro-immune dysfunction in- chemokine levels after antidepressant drug treatment for major depressive disorder:
duced by chronic unpredictable mild stress in the rat, Behav. Brain Res. 356 (2019) systematic review and meta-analysis, Mol. Neurobiol. 55 (2018) 4195–4206,
348–357, https://doi.org/10.1016/j.bbr.2018.07.001. https://doi.org/10.1007/s12035-017-0632-1.
[64] J. Xue, H. Li, X. Deng, Z. Ma, Q. Fu, S. Ma, L-Menthone confers antidepressant-like [79] D.C. Blanchard, K. Meyza, Risk assessment and serotonin: animal models and
effects in an unpredictable chronic mild stress mouse model via NLRP3 in- human psychopathologies, Behav. Brain Res. (2017), https://doi.org/10.1016/j.
flammasome-mediated inflammatory cytokines and central neurotransmitters, bbr.2017.07.008.
Pharmacol. Biochem. Behav. 134 (2015) 42–48, https://doi.org/10.1016/j.pbb. [80] U.E. Lang, S. Borgwardt, Molecular mechanisms of depression: perspectives on new
2015.04.014. treatment strategies, Cell. Physiol. Biochem. 31 (2013) 761–777, https://doi.org/
[65] Y. Song, R. Sun, Z. Ji, X. Li, Q. Fu, S. Ma, Perilla aldehyde attenuates CUMS-induced 10.1159/000350094.
depressive-like behaviors via regulating TXNIP/TRX/NLRP3 pathway in rats, Life [81] P. Xu, K.Z. Wang, C. Lu, L.M. Dong, J. Le Zhai, Y.H. Liao, S. Aibai, Y. Yang, X.M. Liu,
Sci. 206 (2018) 117–124, https://doi.org/10.1016/j.lfs.2018.05.038. Antidepressant-like effects and cognitive enhancement of the total phenols extract
[66] N.A. Siddiqui, N. Ahmad, N. Musthaq, I. Chattopadhyaya, R. Kumria, S. Gupta, of Hemerocallis citrina Baroni in chronic unpredictable mild stress rats and its re-
Neuropharmacological profile of extracts of aerial parts of Convolvulus pluricaulis lated mechanism, J. Ethnopharmacol. 194 (2016) 819–826, https://doi.org/10.
Choisy in mice model, Open Neurol. J. 8 (2014) 11–14, https://doi.org/10.2174/ 1016/j.jep.2016.09.023.
1874205X01408010011. [82] H. mei Jia, M. Yu, L.Y. Ma, H. wu Zhang, Z. mei Zou, Chaihu-Shu-Gan-San regulates
[67] H. Gao, X. Zhu, Y. Xi, Q. Li, Z. Shen, Y. Yang, Anti-depressant-like effect of atrac- phospholipids and bile acid metabolism against hepatic injury induced by chronic
tylenolide I in a mouse model of depression induced by chronic unpredictable mild unpredictable stress in rat, J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 1064
stress, Exp. Ther. Med. (2017) 1574–1579, https://doi.org/10.3892/etm.2017. (2017) 14–21, https://doi.org/10.1016/j.jchromb.2017.08.003.
5517. [83] K.-K. Jia, S.-M. Pan, H. Ding, J.-H. Liu, Y.-J. Zheng, S.-J. Wang, Y. Pan, L.-D. Kong,
[68] V.S. Velingkar, G.L. Gupta, N.B. Hegde, A current update on phytochemistry, Chaihu-shugan san inhibits inflammatory response to improve insulin signaling in
pharmacology and herb–drug interactions of Hypericum perforatum, Phytochem. liver and prefrontal cortex of CUMS rats with glucose intolerance, Biomed.
Rev. 16 (2017) 725–744, https://doi.org/10.1007/s11101-017-9503-7. Pharmacother. 103 (2018) 1415–1428, https://doi.org/10.1016/j.biopha.2018.04.
[69] C. Park, E. Brietzke, J.D. Rosenblat, N. Musial, H. Zuckerman, R.M. Ragguett, 171.
Z. Pan, C. Rong, D. Fus, R.S. McIntyre, Probiotics for the treatment of depressive [84] K.-K. Jia, Y.-J. Zheng, Y.-X. Zhang, J.-H. Liu, R.-Q. Jiao, Y. Pan, L.-D. Kong, Banxia-
symptoms: An anti-inflammatory mechanism? Brain Behav. Immun. 73 (2018) houpu decoction restores glucose intolerance in CUMS rats through improvement of
115–124, https://doi.org/10.1016/j.bbi.2018.07.006. insulin signaling and suppression of NLRP3 inflammasome activation in liver and
[70] N. Lichtblau, F.M. Schmidt, R. Schumann, K.C. Kirkby, H. Himmerich, Cytokines as brain, J. Ethnopharmacol. 209 (2017) 219–229, https://doi.org/10.1016/j.jep.
biomarkers in depressive disorder: current standing and prospects, Int. Rev. 2017.08.004.
Psychiatry 25 (2013) 592–603, https://doi.org/10.3109/09540261.2013.813442.

1708

You might also like