Journal Pre-proof Angiotensin-converting enzyme inhibitor rapidly ameliorates depressive-type behaviors via bradykinin-dependent activation of mTORC1 Han Luo, Peng-Fei Wu, Yu Cao, Ming Jin, Tian-Tian Shen, Ji Wang, Jian-Geng Huang, Qian-Qian Han, Jin-Gang He, Si-Long Deng, Lan Ni, Zhuang-Li Hu, Li-Hong Long, Fang Wang, Jian-Guo Chen PII:
S0006-3223(20)30094-9
DOI:
https://doi.org/10.1016/j.biopsych.2020.02.005
Reference:
BPS 14125
To appear in:
Biological Psychiatry
Received Date: 26 October 2019 Revised Date:
22 January 2020
Accepted Date: 3 February 2020
Please cite this article as: Luo H., Wu P.-F., Cao Y., Jin M., Shen T.-T., Wang J., Huang J.-G., Han Q.Q., He J.-G., Deng S.-L., Ni L., Hu Z.-L., Long L.-H., Wang F. & Chen J.-G., Angiotensin-converting enzyme inhibitor rapidly ameliorates depressive-type behaviors via bradykinin-dependent activation of mTORC1, Biological Psychiatry (2020), doi: https://doi.org/10.1016/j.biopsych.2020.02.005. This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version will undergo additional copyediting, typesetting and review before it is published in its final form, but we are providing this version to give early visibility of the article. Please note that, during the production process, errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain. © 2020 Published by Elsevier Inc on behalf of Society of Biological Psychiatry.
Luo H et al
1
Angiotensin-converting
enzyme
inhibitor
rapidly
ameliorates
2
depressive-type behaviors via bradykinin-dependent activation of
3
mTORC1
4
Han Luo1,*, Peng-Fei Wu1,2,3,4,*, Yu Cao1, Ming Jin6, Tian-Tian Shen1, Ji Wang1,
5
Jian-Geng Huang6, Qian-Qian Han1, Jin-Gang He1, Si-Long Deng1, Lan Ni1,
6
Zhuang-Li Hu1,2,3, Li-Hong Long1,2,3,4, Fang Wang1,2,3,4,5, † and Jian-Guo Chen1,2,3,4,5, †
7
1
8
Huazhong University of Science and Technology, Wuhan City, Hubei 430030, China
9
2
Department of Pharmacology, School of Basic Medicine, Tongji Medical College,
Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China,
10
Wuhan, Wuhan City, Hubei 430030, China
11
3
12
of Hubei Province, Wuhan City, Hubei 430030, China
13
4
14
University of Science and Technology, Wuhan, 430030, China
15
5
The Collaborative-Innovation Center for Brain Science, Wuhan 430030, China
16
6
Department of Pharmaceutics, College of Pharmacy, Tongji Medical College,
17
Huazhong University of Science and Technology, Wuhan, China
18
Short title: ACEI may serve as fast-acting anti-depressants
19
*
The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation
Laboratory of Neuropsychiatric Diseases, The Institute of Brain Research, Huazhong
Equally contributed authors † Correspondence to: Dr. Jian-Guo Chen or Dr. Fang 1
Luo H et al
1
Wang.
Department of Pharmacology, Tongji Medical College, Huazhong University
2
of Science and Technology, 13 Hangkong Road, Wuhan, Hubei, China 430030
3
E-mail:
[email protected] or
[email protected]
4
Tel: +86-27-83692636; FAX: +86-27-83692608
5 6
Key Words: MDD, Angiotensin-converting enzyme inhibitor, Bradykinin, Cdc42,
7
mTORC1 , captopril
8 9
Word count of the abstract:
247
10
Word count of the main text: 3982
11
Number of figures: 6
12
Number of table(s): 0
13
Number of supplement(s): 8
14 15 16 17 18 19 20 21 22 23 24 25 26 27 2
Luo H et al
1
Abstract
2
BACKGROUND: Angiotensin-converting enzyme inhibitors (ACEIs) are widely
3
prescribed anti-hypertensive agents. Intriguingly, case reports and clinical trials have
4
indicated that ACEIs, including captopril and lisinopril, may have a rapid
5
mood-elevating effect in certain patients, but few experimental studies have
6
investigated their value as fast-onset antidepressants. METHODS: The present study
7
consisted of a series of experiments using biochemical assays, immunohistochemistry
8
and behavioral techniques to examine the effect and mechanism of captopril on
9
depressive-like behavior in two animal models, chronic unpredictable stress (CUS)
10
model and chronic social defeat stress (CSDS) model. RESULTS: Captopril (19.5 or
11
39 mg/kg, intraperitoneal injection) exerted rapid antidepressant activity in the
12
CUS-treated and CSDS-treated mice. Pharmacokinetic analysis revealed that captopril
13
crossed the blood brain barrier (BBB) and that lisinopril, another ACEI with better
14
BBB permeability, exerted a faster and longer-lasting effect at a same molar
15
equivalent dose. This antidepressant effect seemed to be independent of the
16
renin-angiotensin system, but dependent on bradykinin (BK) system, since the
17
decreased BK detected in the stressed mice could be reversed by captopril. The
18
hypofunction of the downstream effector of BK, cell division control protein 42
19
homolog, contributed to the stress-induced loss of dendritic spines, which was rapidly
20
reversed by captopril via activating the mammalian target of rapamycin complex 1
21
(mTORC1) pathway. CONCLUSIONS: Our findings indicate that the BK-dependent 3
Luo H et al
1
activation of mTORC1 may represent a promising mechanism underlying
2
antidepressant pharmacology. Considering their affordability and availability, ACEIs
3
may emerge as a novel fast-onset antidepressant, especially for patients with
4
comorbid depression and hypertension.
5
6
Introduction
7
Major depressive disorder (MDD) is a significant contributor to the global burden
8
of disease and affects approximately 16% of the world’s population at some point in
9
their lives. The major problems in the therapy of MDD are that only 40-70% of
10
patients with depression respond to drug treatment, and that the onset of the
11
therapeutic effect is in a delayed manner. In recent years, there has been substantial
12
clinical and preclinical progress in identifying fast-onset antidepressants, such as
13
ketamine and scopolamine (1-3), and the enantiomer S-ketamine has been recently
14
approved to be prescribed for treatment-resistant depression by the U.S. food and drug
15
administration (FDA). However, the use of available fast-onset antidepressants is
16
limited due to their risk of dependence and psychomimetic side effects.
17
Some
clinic
studies
have
revealed
a
possible
relationship
between
18
cerebrovascular disease and occurrence or outcomes of depression in later life (4-6).
19
Angiotensin II (Ang II), the most important component of renin-angiotensin-system
20
(RAS),
is
assumed
to
stimulate
the
hyperactivity
of
the 4
Luo H et al
1
hypothalamic-pituitary-adrenocortical axis via activation of Ang II type 1 receptor
2
(AT1R) in corticotropin-releasing-hormone neurons (7-8). Clinical data indicate that
3
RAS-acting agents, including AT1R blocker (ARB) and angiotensin-converting
4
enzyme (ACE) inhibitor, reduce the risk of mood disorders compared with the
5
patients taking other antihypertensive drugs such as calcium channel blockers and
6
β-blockers (9). Intriguingly, although no randomized controlled trial has assessed the
7
effects, in the last forty years, a succession of case reports and clinical studies have
8
reported that ACEIs, not ARBs, elicit mood-elevating effects in certain hypertensive
9
patients (10-17), which were indicated by the time axis in Fig.S1. In the early 1980s,
10
several cases reported that captopril might rapidly improve the patient's mood within
11
1-2 days at the daily dosages ranging from 37.5 - 200 mg (10-11, 13). In 2005, mood
12
benefits were observed in 9 hypertensive patients with MDD, who were treated with
13
another ACEI, lisinopril (18). ACEIs are a group of widely prescribed
14
anti-hypertensive agents, but to date, very few experimental studies have investigated
15
their antidepressant value (19-20). In most hypertensive patients, the daily dosage of
16
captopril is 12.5 - 150 mg (a single dosage is 12.5 - 50 mg, one to three times per day).
17
Thus, we screened the effects of captopril on depressive-like behaviors in mice as
18
following: 2.44, 4.88, 9.75, 19.5 and 39 mg/kg/day (the equivalent of a single dosage
19
of 11.87, 23.74, 47.48, 94.96 or 189.92 mg in a human weighing 60 kg), using the
20
body surface area normalization method described by FDA draft guidelines (21,
21
human dosage = mice dosage × Km mice /Km human, Km mice = 3, Km human = 5
Luo H et al
1
37). We found that only high dosage of captopril produced a fast-onset therapeutic
2
effect on depressive-like behaviors.
3
An increasing number of studies have demonstrated that activation of mammalian
4
target of rapamycin (mTOR) signaling in the medial prefrontal cortex (mPFC)
5
mediate the rapid antidepressant actions of ketamine (3, 22). A recent report has
6
indicated that direct activation of mTOR signaling via a leucine sensing pathway by
7
the sestrin modulator NV-5138 mimics the rapid antidepressant effects of ketamine
8
(23). It has been reported that ACE may block mTOR signaling pathway (24-25), but
9
very little is known about the downstream signaling mechanism. ACE function
10
comprises the production of Ang II and the breakdown of bradykinin (BK). Ang II,
11
the main product of ACE, increases the mTOR activity (26-27), whereas BK, a
12
degraded substrate for ACE, also activates mTOR signaling pathway via B2 receptor
13
(B2R) in various conditions (28-30). We further determined that ACEI may work by
14
activating the mammalian target of rapamycin complex 1 (mTORC1) pathway via a
15
non-RAS mechanism, i.e., a BK-dependent pathway, and identified B2R as a novel
16
therapeutic target for depression.
17
18
Materials and Methods
19
Detailed Materials and Methods are available in Supplementary Materials and
20
Methods.
6
Luo H et al
1
2
Animals and behavioral experiments. Male C57BL/6J mice (7-8 weeks of age,
3
18-21 g) from Hunan SJA Laboratory Animal (Changsha, Hunan, China) were used in
4
our study. All the procedures were conducted following the Declaration of Helsinki
5
and the Guide for Care and Use of Laboratory Animals as adopted and promulgated
6
by the National Institutes of Health. All experiments were approved by the Review
7
Committee for the Use of Human or Animal Subjects of Huazhong University of
8
Science and Technology. Chronic unpredictable stress (CUS) and chronic social
9
defeat stress (CSDS) were used to induce depressive-like behavior and all the
10
behavioral tests were conducted as previously described with slight modifications
11
(31-33). Sample sizes were determined according to those used in previous
12
publications from our group and others similar studies (31, 34-35) and justified by the
13
power analyses.
14
15
Experiments in molecular biology. Quantitative real-time PCR (qPCR) was
16
performed on the StepOnePlusTM Real-Time PCR System (Applied Biosystems,
17
Foster City, CA) to analyze the gene expression. Western blotting was used to analyze
18
the protein level. The vectors contained a cytomegalovirus-driven enhanced green
19
fluorescent protein (EGFP) and oligonucleotides encoding short hairpin RNAs
20
(shRNAs) of B2R were purchased from Shanghai Genechem Co., Ltd. (Shanghai,
21
China). The assay of cell division control protein 42 homolog (Cdc42) activity was 7
Luo H et al
1
performed by analyzing GTP-Cdc42/Cdc42 ratio as previous reports (36).
2
3
Statistics. Analysis was performed using Graphpad Prism 7.0 or SPSS 18.0 software
4
(SPSS Inc., Chicago, IL, USA) and p < 0.05 was considered statistically significant.
5
All values were expressed as mean ± S.E.M. Each “n” corresponded to a single mouse.
6
If technical replicates were performed, their mean was considered as one “n”. We
7
tested the data normality using Kolmogorov-Smirnov test of normality with the
8
Dallal-Wilkinson-Lillie corrected P value (GraphPad Prism 7.0), and variances were
9
compared by Bartlett statistics to decide whether parametric tests were applicable.
10
Statistical analyses were performed using one-way ANOVA followed by LSD
11
multiple comparison tests or two-way ANOVA followed by Bonferroni test to
12
compare means of three or more groups, one-way repeated measures ANOVA to
13
examine means of repeated measured data and unpaired two-tailed Student’s t-test to
14
compare two groups. For the non-normal distributed data (data for AT1R in the mPFC
15
in Fig.3A, data for CUS and CUS + Captopril group in Fig.3B), Mann-Whitney test
16
were used.
17
18
Results
19
Captopril rapidly reverses chronic stress-induced depressive-like behaviors in
20
mice. 8
Luo H et al
1
First, the effects of captopril on behavioral despair were measured by immobility
2
time in two behavioral tests, the tail suspension test (TST) and forced swimming test
3
(FST). A wide range of clinically relevant dosages of captopril were chosen (2.44,
4
4.88, 9.75, 19.5, and 39 mg/kg/day in mice). Intraperitoneal injection of captopril at
5
dosages of 9.75, 19.5 and 39 mg/kg/day produced rapid antidepressant responses in
6
the FST and TST 24 h after administration (Fig. 1A, B). However, captopril exhibited
7
few influences on the open field test (OFT, Fig. S2A-C), elevated plus maze (EPM,
8
Fig. S2D-F) and the novelty-suppressed feeding test (NSFT, Fig. S2G), indicating that
9
captopril may exert little effect on anxiety. At the same dosage (19.5 or 39 mg/kg, i.p),
10
captopril exerted little effect on water drinking (12 h, Fig. S2H). The effect of
11
captopril in the FST was detected within 24 h and lasted for 7 days after one dosing
12
(Fig. S2I). The levels of captopril in the mPFC were measured using liquid
13
chromatography-tandem mass spectrometry (LC-MS/MS) following the peripheral
14
administration of captopril (19.5 mg/kg, Fig. 1C). The plasma concentrations of
15
captopril reached peak levels (184.06 ± 20.25 ng/ml) at 0.5 h after administration. In
16
this experimental condition, the detection limit of captopril concentration was 23.01
17
nM (5 ng/ml). A modest concentration of captopril was detected in the mPFC tissue
18
(24.6 ± 2.82 ng/ml) 0.5 h after administration, indicating that captopril can enter the
19
blood brain barrier (BBB) at high dosage.
20
Next, the mice that exposed to CUS were used to evaluate the effects of captopril.
21
CUS mice displayed anhedonia, a core symptom of depression measured by the SPT, 9
Luo H et al
1
increased despair behaviors and a reduction in body weight (Fig. S3A-E). Notably, the
2
CUS-induced reduction in sucrose preference was reported to be reversed by daily
3
treatment with tricyclic antidepressants for 2 to 3 weeks (37-38); however, it was
4
rapidly reversed by a single dosage (19.5 or 39 mg/kg, Fig. 1D) of captopril within 24
5
h. This effect was not due to the effect of angiotensin on drinking behavior because
6
the CUS mice treated with captopril (0, 19.5 or 39 mg/kg, i.p) displayed similar total
7
fluid consumption in the SPT (n = 8-12, Fig. S3F). Moreover, the increased sucrose
8
preference was observed at 1, 3 and 7 days after interperitoneal injection of captopril
9
(19.5 mg/kg), indicating that the antidepressant effects of captopril on anhedonia can
10
last for at least 1 week (Fig. 1E). These results consistently demonstrate that captopril
11
elicits fast and sustained antidepressant effects in mice.
12
We also employed another animal model of depression, CSDS, to evaluate the
13
effect of captopril on social deficits. We found that the social index increased at 24 h
14
after intraperitoneal injection of captopril (19.5 mg/kg, Fig. 1F), indicating that
15
captopril improved the social interaction in the socially defeated mice. The effects of
16
a single dosage of captopril on blood pressure and locomotor activity in CUS mice
17
were very limited 24 h after administration (Fig. S4), indicating that its rapid
18
antidepressant activity may not be associated with the alterations in blood pressure.
19
20
Other RAS-acting agents are insufficient to elicit rapid antidepressant effects. 10
Luo H et al
1
The Ang II reducing effect largely mediates the cardiovascular effects of ACEI.
2
Thus, we investigated whether other RAS-acting agents mimicked the rapid
3
antidepressant activity of ACEI. First, we employed a direct renin inhibitor (DRI),
4
aliskiren, to mimic the Ang II-reducing effect of captopril (Fig. 2A). Aliskiren (10
5
mg/kg, 50 mg/kg) induced a significant reduction in Ang II levels 24 h after
6
administration (Fig. S5A), but failed to exert significant antidepressant effects in the
7
CUS-treated mice (Fig. 2B), indicating that Ang II-reducing effect may be insufficient
8
to elicit rapid antidepressant effects. Then, valsartan, a specific and widely used ARB,
9
was applied to mimic the captopril-mediated hypofunction of AT1R. A previous
10
report found that pretreatment with losartan (20 mg/kg), another ARB, reduced the
11
duration of immobility in the FST of normal normal male CD mice (39). However, in
12
the CUS mice, the intraperitoneal injection of valsartan (30 or 60 mg/kg) elicited no
13
observed rapid antidepressant effects (Fig. 2C), which was consistent with a very
14
recent report performed on CUS-treated rats (40). We also administered an Ang II
15
neutralizing antibody (nAb), aliskiren and valsartan directly into the mPFC, again,
16
and they did not exert rapid antidepressant effects (Fig. 2D and Fig. S5B), suggesting
17
that down-regulation of Ang II function in either the peripheral or central, may be
18
insufficient to produce rapid antidepressant effects in the stressed mice.
19
Then, we asked whether other ACEIs have similar antidepressant effects and
20
compared the efficacy/sustainability of lisinopril, a long-lasting ACEI that can pass
21
through the BBB (41), with that of captopril. At a molar equivalent dosage to that of 11
Luo H et al
1
captopril (19.5 mg/kg), the duration of the antidepressant effect of lisinopril (39.6
2
mg/kg) was much longer than that of captopril in non-stressed mice (Fig. 2E). In the
3
stressed mice, lisinopril (39.6 mg/kg) exerted a much faster antidepressant effect
4
(within two hours) than that of captopril (Fig. 2F).
5
6
Reactivation of the BK system mediates the rapid antidepressant effects of
7
ACEI.
8
We observed that the concentration of Ang II, as well as the mRNA levels of
9
angiotensinogen (AGT), ACE, AT1R and Ang II type 2 receptor (AT2R), exhibited
10
few changes in the mPFC, hippocampus and nucleus accumbens (NAc) of the
11
CUS-treated mice (Fig. 3A). As a vasodilator nonapeptide that is degraded by ACE,
12
BK is another important molecular mediator underlying the cardiovascular effects of
13
ACEI as a non-RAS mechanism. Interestingly, the levels of BK were significantly
14
decreased in the mPFC and plasma of stressed mice (Fig. 3B-D), an effect that was
15
reversed by captopril (19.5 mg/kg, i.p.). To assess the relevance of BK levels to MDD,
16
we analyzed the BK concentration in the plasma of human subjects who were
17
diagnosed with MDD. Notably, a similar change in BK levels was observed in the
18
plasma of depressed patients (Fig. 3E), as determined by ELISA. BK exerts its effects
19
via two different receptor subtypes: B1 receptor (B1R) and B2R. The level of B2R,
20
not B1R in the mPFC of CUS mice was up-regulated (Fig. 3F). The altered expression 12
Luo H et al
1
of B2R may confer a compensatory mechanism of BK deficits induced by CUS.
2
Previous reports have indicated that when administered in the central nervous
3
system (CNS), BK leads to initial rapid excitation (42) and hyperalgesia (43). Thus,
4
we asked whether the BK system mediated the rapid antidepressant effect of captopril.
5
To address this issue, BK (50 ng/per side) was bilaterally infused into the mPFC of
6
stressed mice to mimic the BK-potentiating property of captopril. We found that the
7
local administration of BK in the mPFC rapidly reversed depressive-like behaviors in
8
the stressed mice (Fig. 3G). Next, we explored whether the inhibition of BK function
9
was associated with depressive-like behaviors. Repeated administration of HOE140
10
(65.226 µg/kg per day, i.p), a blocker of B2R, but not DALBK (99.818 µg/kg per day,
11
i.p), a blocker of B1R, for 7 days significantly increased the immobility time in the
12
FST of mice (Fig. 3H), suggesting that hypofunction of B2R may contribute to the
13
pathophysiology of depression. Together, these results demonstrate that B2R
14
signaling may play a key role in the antidepressant activity of captopril.
15
To further confirm the role of B2R in the antidepressant mechanism of captopril,
16
we employed both pharmacological and genetic approaches in this study. Intra-mPFC
17
infusion of HOE140 (100 nM, 1 µl per side), but not DALBK (100 nM, 1 µl per side),
18
completely blocked captopril-induced reduction in the immobility time of TST and
19
FST (Fig. 4A). We further examined the influence of HOE140 on the antidepressant
20
effect of captopril in the CUS-treated mice. Intra-mPFC injection of HOE140
21
abolished the behavioral responses to captopril in stressed mice (Fig. 4B). To directly 13
Luo H et al
1
explore the role of B2R in the effects of captopril, we used a lentivirus that expressed
2
short hairpin RNAs to knockdown B2R expression. As shown in Fig.S6, the
3
lentivirus-guided EGFP expression was predominantly located in the mPFC and the
4
B2R protein level was significantly down-regulated by LV-B2R siRNA. We found
5
that captopril failed to exert an antidepressant effect in the mice with lentivirus-guided
6
knockdown of B2R (Fig.S6 and Fig. 4C). These results demonstrate that BK-B2R
7
signaling determines the antidepressant effect of captopril.
8
9 10
BK-stimulated Cdc42 activity confers the antidepressant effects of captopril via activation of the mTORC1 pathway.
11
B2R is widely distributed in the CNS (44) and a previous study has revealed that
12
BK depolarizes motor neurons by postsynaptic activation of B2R (45). We
13
hypothesized that B2R may initiate the antidepressant activity via postsynaptic action.
14
BK is a powerful stimulator of Cdc42, a critical Rho GTPase protein (46-47).
15
Growing evidence suggests that Cdc42 controls rapid presynaptic maturation to
16
facilitate synaptogenesis (48) and also contributes to postsynaptic maturation (49).
17
Furthermore, Cdc42 regulates the activation of the mTORC1 signaling pathway
18
(50-51), which is critical in the synaptic mechanisms underlying rapid-acting
19
antidepressants. Thus, we asked whether the Cdc42-mTOR signaling pathway
20
contributes to the antidepressant activity of captopril. 14
Luo H et al
1
First, we found that the activity of Cdc42 was reduced significantly in the mPFC
2
of CUS-treated mice, and captopril rapidly rescued the decrease in Cdc42 activity in
3
the mPFC (Fig. 5A). As shown in Fig. 5B & C, the intra-mPFC infusion of a selective
4
Cdc42 inhibitor ML141 (8.15 mg/µl/side) blocked the effect of captopril on despair
5
behaviors in the non-stressed mice (Fig. 5B), and the pre-infusion of ML141 into the
6
mPFC abolished the antidepressant activity of captopril in the stressed mice (Fig. 5C).
7
These results suggest that Cdc42 plays a role in the mechanism in the antidepressant
8
activity of captopril.
9
We next examined the effect of captopril on the mTORC1 activity. The
10
phosphorylated forms of mTORC1 (p-mTOR) and the key downstream target of
11
mTORC1, p70S6 kinase (p-p70S6K) represent the activation of mTORC1 signaling.
12
We found that captopril activated mTORC1 in the non-stressed mice, moreover, the
13
intra-mPFC infusion of ML141 prevented the captopril-induced activation of
14
mTORC1 (Fig. 5D). In the CUS-treated mice, captopril reversed the stress-induced
15
inhibition of mTORC1 activity (Fig. 5E, p-mTOR and p-p70S6K) and induced an
16
increased level of BDNF (Fig. 5E). The genetic knockdown of B2R abolished the
17
captopril-induced activation of mTORC1 and BDNF synthesis in the stressed mice
18
(Fig. 5E). We next assessed whether increased mTOR activity is sufficient to mediate
19
the rapid antidepressant activity of captopril. Accordingly, a selective mTORC1
20
inhibitor, rapamycin, was infused into the mPFC of CUS mice. We found that the
21
infusion of rapamycin into the mPFC abolished captopril-elicited rapid antidepressant 15
Luo H et al
1
activity (Fig. 5F).
2
To further address the relationship between BK and mTORC1 activity, we
3
observed a direct action of BK on mTORC1 activity in primary cultured neurons.
4
Incubation of BK (10 nM) increased phosphorylation of p70S6K and BDNF level in
5
primary cultured neurons of mPFC (Fig. S7A-B). Additionally, in vivo, we found that
6
rapamycin blocked BK-elicited rapid antidepressant responses (Fig. S7C). Thus,
7
captopril may exert antidepressant effects through the BK-B2R-Cdc42-mTORC1
8
signaling pathway.
9
10
Captopril reverses CUS-induced synaptic loss and stimulates synaptogenesis.
11
Considering that Cdc42 activity can regulate dendritic spine plasticity, we asked
12
whether the reactivation of Cdc42 function can alleviate the CUS-induced synaptic
13
loss. Using confocal microscopy, the dendritic spine density in the mPFC of stressed
14
mice was measured 24 h after captopril administration. As previously reported (52),
15
dendritic spines were classified by functional subtype: long thin, mushroom and
16
stubby. In our study, the CUS exposure significantly reduced the number and density
17
of dendritic spines, especially long thin spines, in the mPFC of mice. Moreover,
18
captopril rapidly rescued the synaptic loss observed in the mPFC of CUS-treated mice
19
by increasing the spine density and number of long thin and mushroom dendritic
20
spines (Fig. 6A-B). The morphological changes were abolished by the intra-mPFC 16
Luo H et al
1
infusion of ML141 (Fig. 6A-B), indicating that Cdc42-dependent synaptogenesis
2
mediates the effect of captopril on CUS-induced synaptic loss. Furthermore, captopril
3
significantly increased the levels of key synaptic proteins, including GluA1 and
4
PSD95, in the mPFC of CUS mice (Fig. 6C), which strengthens our hypothesis that
5
captopril exerts antidepressant effects by increasing synapse numbers. Captopril also
6
increased the levels of synaptic proteins, including GluA1 and PSD95 in the
7
hippocampus of CUS mice (Fig. 6D), suggesting that in addition to the mPFC, other
8
brain areas may be involved in the effect of captopril.
9
10
Discussion
11
In the present study, we demonstrated that ACEIs produced a rapid and
12
long-lasting reversal of chronic stress-induced depressive-like behaviors by
13
potentiating the BK-B2R-Cdc42-mTORC1 signaling pathway (Fig. 6E). Our study
14
proposed a new property of ACEI, an important class of RAS-acting agents. As
15
widely used anti-hypertensive agents, the clinical antidepressant value of RAS-acting
16
agents has not been established. Our results indicated that RAS-acting agents
17
including DRI, ARB and captopril at a single clinic dosage, did not exert a rapid
18
antidepressant activity in CUS-treated rodents. Interestingly, our data indicated that
19
the BK action, an acute physiological outcome that was often limited by the clinical
20
dosage regimen to avoid side effects, mediated the antidepressant effects of ACEIs at 17
Luo H et al
1
high dosages. An alternative interpretation for the overlook of the ACEI effect may be
2
that captopril initiated a rapid but transient action for a few days, like ketamine, which
3
was approved for anesthetic in 1970, but its antidepressant value has been established
4
until recent years.
5
BK plays a key role in the pharmacological effect of captopril (53). A notable
6
finding of the present study is that the altered BK system may contribute to the
7
development of depressive-like behaviors. Decreased levels of BK in the blood and
8
mPFC were observed in mice exposed to CUS. The administration of BK in the
9
mPFC rapidly rescued CUS-induced behavior deficits. Consistently, the levels of BK
10
were also decreased in the plasma of MDD patients. It should be noted that the
11
differences in the gender composition of human samples may induce bias. We
12
analyzed the bradykinin level between wemen and men, and no differences were
13
found, both in the MDD patients and healthy volunteers. However, considering the
14
sex ratio in control population does not mirror the ratio in the experimental group, and
15
all the experiments were performed on the male mice, the role of bradykinin in
16
women depressive patients should be further evaluated.
17
Increased B2R levels in the mPFC of stressed mice were observed, and these
18
increased levels may be a compensatory mechanism in response to chronic
19
stress-induced deficits in BK signaling. We found that repeated administration of the
20
B2R blocker HOE140 (65.226 µg/kg per day, i.p) for 7 days increased FST
21
immobility time (seen in Fig.3H). However, in the Fig.4A, intra-mPFC infusion of 18
Luo H et al
1
HOE140 (100 nM, 1 µl per side, once) did not affect the immobility time after 24
2
hours, indicating that a long-term blockade, not acute blockade of B2R, may generate
3
despair behaviors. We hypothesized that chronic blockade of B2R in the PFC would
4
also have increased immobility, and this point was supported by our data that the
5
genetic knockdown of B2R in the mPFC increased FST immobility time in control
6
mice (Fig.4C). Both pharmacological and genetic approaches revealed that B2R, not
7
B1R, mediated the antidepressant effects of BK and captopril. Considering that a
8
neuroprotective role of B2R has been revealed (54), our findings raise the possibility
9
that BK-potentiating peptides or drugs may emerge as a new class of antidepressants.
10
Notably, BK-induced cough and hyperalgesia may affect treatment compliance,
11
however, it could be prevented by a rational drug regimen or a peripheral blocker of
12
BK receptors.
13
Previous studies have reported that BK potentiates synaptic transmission via
14
activating both presynaptic and postsynaptic B2Rs (45, 55). As a G-protein-coupled
15
receptor, B2R strongly stimulates Cdc42 activation (46-47). It is generally believed
16
that the Cdc42 signaling pathway plays a key role in the structural plasticity of
17
dendritic spines, dendritic morphogenesis, synaptic maturation and axon guidance
18
(56). MDD has been linked to aberrant dendritic spine and synapse development (57).
19
Until now, very little is known about the role of Cdc42 in depression. In our study,
20
we found that CUS induced a robust defect in Cdc42 activation in the mPFC, which
21
could be rescued by captopril. Meanwhile, captopril restored CUS-induced spine loss. 19
Luo H et al
1
Both the behavior and morphologic effects of captopril were abolished by the Cdc42
2
inhibitor ML141, suggesting that a Cdc42-dependent mechanism may be critical to
3
the effect of captopril.
4
In the recent decade, ketamine and scopolamine have been developed as
5
rapid-acting antidepressants that can improve depressive symptoms within hours or
6
days in patients. The promotion of mTORC1 has been recognized as a common
7
signaling pathway that mediates rapid-acting antidepressant effects. A recent report
8
indicated that direct activation of mTORC1 via a leucine sensing pathway by
9
NV-5138 mimicked the rapid antidepressant effects of ketamine without affecting
10
glutamate receptors (23), and Navitor Pharmaceuticals has commenced Phase I
11
Clinical Evaluation of NV-5138 in patients with treatment-resistant depression.
12
Similarly, we found that captopril activated the mTORC1 pathway via a
13
BK-dependent pathway. BK incubation directly activated the mTOR signaling in the
14
cultured neurons (Fig. S7A-B), which may work through a BK-B2R-Cdc42-mTORC1
15
signaling pathway. Previous study has reported that, in an in vitro study,
16
overexpression of ACE reduced the level of p70S6K, whereas captopril increased the
17
levels of p70S6K (24), which was in consistent with our observations. We found that
18
captopril promoted synaptogenesis, which may be due to that captopril and BK,
19
similar to other rapid-acting antidepressants, facilitated the mTORC1-dependent
20
synaptogenesis.
21
ACEIs are widely used antihypertensive agents, and some studies also indicated 20
Luo H et al
1
ACEIs might be beneficial for psychiatric diseases (58). Considering the affordability
2
and availability of ACEIs, long-lasting ACEIs that can cross the BBB may be used as
3
new rapid-onset antidepressants. The pharmacodynamics and toxicity of ACEIs have
4
been well studied. Considering the fetal toxicity, and the increase of neonatal
5
morbidity and death, the use of ACEI for depression during pregnancy should be
6
limited. In patients with comorbid depression and diabetes or impaired renal function,
7
ACEI should be used with caution. A further large-scale, randomized, controlled
8
clinical study should be performed to evaluate the antidepressant effect of ACEIs.
9
10
Acknowledgments
11
This work was supported by grants from the Foundation for Innovative Research
12
Groups of NSFC (No. 81721005 to J.G.C. and F. W.), National Natural Science
13
Foundation of China (No. 81773712 to P.F.W., No. 81471377 and No. 81671438 to
14
F.W., No. 81473198 and No. 81673414 to J.G.C.), PCSIRT (No. IRT13016) to J.G.C.,
15
the Program for HUST Academic Frontier Youth Team and Integrated Innovative
16
Team for Major Human Diseases Program of Tongji Medical College, HUST, to F. W.
17
18
Author contributions
19
H.L. performed most molecular and behavioral experiments, stereotaxic surgeries and
21
Luo H et al
1
analyzed data. Peng-Fei Wu designed the experiments, performed molecular
2
experiments, helped methodology and analyzed data. Y.C., Q.Q.H. and S.L.D.
3
contributed to animal experiments and stereotaxic surgeries. M.J and J.G. H
4
performed LC-MS/MS analysis of captopril. T.T.S. performed CSDS model and
5
contributed to behavioral experiments. J.W. and L.N. contributed to confocal
6
microscopy experiments. J.G.H. contributed to measure plasma BK content. Z.L.H.
7
and L.H.L. provided the technique supports. F.W. and J.G.C. supervised the project,
8
designed the experiments, revised the manuscript and supported funding acquisition.
9
P.F.W., H.L., F.W. and J.G.C. wrote the paper with contributions from all of the other
10
authors.
11
12
Financial Disclosures: The authors report no biomedical financial interests or
13
potential conflicts of interest.
14 15
Reference
16
1.
17
models. Mol Psychiatr. 22:(5(-((5.
18
2.
19
synapse formation underlies the rapid antidepressant effects of NMDA antagonists. Science.
20
329:959-9(4.
Ramaker MJ, Dulawa SC (2017–: Identifying fast-onset antidepressants using rodent
Li N, Lee B, Liu RJ, Banasr M, Dwyer JM, Iwata M, et al. (2010–: mTOR-dependent
22
Luo H et al
Voleti B, Navarria A, Liu RJ, Banasr M, Li N, Terwilliger R, et al. (2013–: Scopolamine
1
3.
2
rapidly increases mammalian target of rapamycin complex 1 signaling, synaptogenesis, and
3
antidepressant behavioral responses. Biol Psychiatry. 74:742-749.
4
4.
5
evolution of depressive symptoms in the Cardiovascular Health Study. Stroke, 33: 1(3(-1(44
6
5.
7
Carotid artery stiffness and incident depressive symptoms: The Paris Prospective Study III.
8
Biol Psychiatry, 85: 498-505.
9
(.
Steffens DC, Krishnan KR, Crump C, Burke GL. (2002–: Cerebrovascular disease and
van Sloten TT, Boutouyrie P, Tafflet M, Offredo L, Thomas F, Guibout C, et al. (2019–:
Steffens DC (2019–: Vascular Depression: Is an Old Research Construct Finally Ready for
10
Clinical Prime Time? Biol Psychiatry, 85:441-442.
11
7.
12
corticotropin-releasing-hormone neurons by angiotensin II. Neuroendocrinology. (1:437-444.
13
8.
14
Polymorphisms in the angiotensin-converting enzyme gene are associated with unipolar
15
depression, ACE activity and hypercortisolism. Mol Psychiatry. 11:1003-1015.
16
9.
17
Monotherapy With Major Antihypertensive Drug Classes and Risk of Hospital Admissions for
18
Mood Disorders. Hypertension. (8:1132-1138.
19
10. Zubenko GS, Nixon RA (1984–: Mood-elevating effect of captopril in depressed patients.
Aguilera G, Young WS, Kiss A, Bathia A (1995–: Direct regulation of hypothalamic
Baghai TC, Binder EB, Schule C, Salyakina D, Eser D, Lucae S, et al. (200(–:
Boal AH, Smith DJ, McCallum L, Muir S, Touyz RM, Dominiczak AF, et al. (201(–:
23
Luo H et al
1
Am J Psychiatry. 141:110-111.
2
11. Deicken RF (198(–: Captopril treatment of depression. Biol Psychiatry. 21:1425-1428.
3
12. Croog SH, Levine S, Testa MA, Brown B, Bulpitt CJ, Jenkins CD, et al. (198(–: The
4
effects of antihypertensive therapy on the quality of life. N Engl J Med. 314:1(57-1((4.
5
13. Germain L, Chouinard G (1988–: Treatment of recurrent unipolar major depression with
6
captopril. Biol Psychiatry. 23:(37-(41.
7
14. Cohen BM, Zubenko GS (1988–: Captopril in the treatment of recurrent major depression.
8
J Clin Psychopharmacol. 8:143-144.
9
15. Vuckovic A, Cohen BM, Zubenko GS (1991–: The use of captopril in treatment-resistant
10
depression: an open trial. J Clin Psychopharmacol. 11:395-39(.
11
1(. Michalsen A, Wenzel RR, Mayer C, Broer M, Philipp T, Dobos GJ (2001–: [Quality of life
12
and psychosocial factors during treatment with antihypertensive drugs. A comparison of
13
captopril and quinapril in geriatric patients]. Herz. 2(:4(8-47(.
14
17. Braszko JJ, Karwowska-Polecka W, Halicka D, Gard PR (2003–: Captopril and enalapril
15
improve cognition and depressed mood in hypertensive patients. J Basic Clin Physiol
16
Pharmacol. 14:323-343.
17
18. Hertzman M, Adler LW, Arling B, Kern M (2005–: Lisinopril may augment antidepressant
18
response. J Clin Psychopharmacol. 25:(18-(20.
19
19. Giardina WJ, Ebert DM (1989–: Positive effects of captopril in the behavioral despair swim 24
Luo H et al
1
test. Biol Psychiatry. 25:(97-702.
2
20. Martin P, Massol J, Puech AJ (1990–: Captopril as an antidepressant? Effects on the
3
learned helplessness paradigm in rats. Biological psychiatry. 27:9(8-974.
4
21. Nair A, Morsy MA, Jacob S (2018–: Dose translation between laboratory animals and
5
human in preclinical and clinical phases of drug development. Drug Develop Res. 79:373-382.
6
22. Li N, Liu RJ, Dwyer JM, Banasr M, Lee B, Son H, et al. (2011–: Glutamate
7
N-methyl-D-aspartate receptor antagonists rapidly reverse behavioral and synaptic deficits
8
caused by chronic stress exposure. Biol Psychiatry. (9:754-7(1.
9
23. Kato T, Pothula S, Liu RJ, Duman CH, Terwilliger R, Vlasuk GP, et al. (2019–: Sestrin
10
modulator NV-5138 produces rapid antidepressant effects via direct mTORC1 activation. J
11
Clin Invest. 129:2542-2554.
12
24. Mori S, Tokuyama K (2007–: ACE activity affects myogenic differentiation via mTOR
13
signaling. Biochem Biophys Res Commun. 3(3:597-(02.
14
25. Chen LJ, Xu YL, Song B, Yu HM, Oudit GY, Xu R, et al. (201(–: Angiotensin-converting
15
enzyme 2 ameliorates renal fibrosis by blocking the activation of mTOR/ERK signaling in
16
apolipoprotein E-deficient mice. Peptides. 79:49-57.
17
2(. Hafizi S, Wang X, Chester AH, Yacoub MH, Proud CG (2004–: ANG II activates effectors
18
of mTOR via PI3-K signaling in human coronary smooth muscle cells. Am J Physiol Heart Circ
19
Physiol. 287:H1232-1238.
25
Luo H et al
1
27. Wang RH, He JP, Su ML, Luo J, Xu M, Du XD, et al. (2013–: The orphan receptor TR3
2
participates in angiotensin II-induced cardiac hypertrophy by controlling mTOR signalling.
3
Embo Mol Med. 5:137-148.
4
28. Fu C, Cao Y, Li B, Xu R, Sun Y, Yao Y (2019–: Bradykinin protects cardiac c-kit positive
5
cells from high-glucose-induced senescence through B2 receptor signaling pathway. J Cell
6
Biochem.
7
29. Liu YP, Lu ZY, Cui M, Yang Q, Tang YP, Dong Q (201(–: Tissue kallikrein protects
8
SH-SY5Y neuronal cells against oxygen and glucose deprivation-induced injury through
9
bradykinin B2 receptor-dependent regulation of autophagy induction. J Neurochem.
10
139:208-220.
11
30. Yu HS, Wang SW, Chang AC, Tai HC, Yeh HI, Lin YM, et al. (2014–: Bradykinin promotes
12
vascular endothelial growth factor expression and increases angiogenesis in human prostate
13
cancer cells. Biochem Pharmacol. 87:243-253.
14
31. Li MX, Zheng HL, Luo Y, He JG, Wang W, Han J, et al. (2018–: Gene deficiency and
15
pharmacological inhibition of caspase-1 confers resilience to chronic social defeat stress via
16
regulating the stability of surface AMPARs. Mol Psychiatry. 23:55(-5(8.
17
32.
18
Protein 150 and Protein Kinase A Complex in the Basolateral Amygdala Contributes to
19
Depressive-like Behaviors Induced by Chronic Restraint Stress. Biol Psychiatry. 8(:131-142.
20
33. Fogaca MV, Campos AC, Coelho LD, Duman RS, Guimaraes FS (2018–: The anxiolytic
Zhou HY, He JG, Hu ZL, Xue SG, Xu JF, Cui QQ, et al. (2019–: A-Kinase Anchoring
26
Luo H et al
1
effects of cannabidiol in chronically stressed mice are mediated by the endocannabinoid
2
system: Role of neurogenesis and dendritic remodeling. Neuropharmacology. 135:22-33.
3
34.
4
ventral tegmental area regulate behavioral responses to chronic stress. Elife. 7: e32420.
5
35.
6
Antidepressant effects of Fibroblast Growth Factor-2 in behavioral and cellular models of
7
depression. Biol Psychiatry. 72: 258 2(5.
8
3(. Benard V, Bohl BP, Bokoch GM (1999–: Characterization of Rac and Cdc42 activation in
9
chemoattractant-stimulated human neutrophils using a novel assay for active GTPases.
Zhong P, Vickstrom CR, Liu X, Hu Y, Yu L, Yu HG, et al. 2018
: HCN2 channels in the
Elsayed M, Banasr M, Duric V, Fournier NM, Licznerski P, Duman RS (2012–:
10
Journal of Biological Chemistry. 274:13198-13204.
11
37. Willner P, Towell A, Sampson D, Sophokleous S, Muscat R (1987–: Reduction of sucrose
12
preference by chronic unpredictable mild stress, and its restoration by a tricyclic
13
antidepressant. Psychopharmacology (Berl–. 93:358-3(4.
14
38. Navarria A, Wohleb ES, Voleti B, Ota KT, Dutheil S, Lepack AE, et al. (2015–: Rapid
15
antidepressant actions of scopolamine: Role of medial prefrontal cortex and M1-subtype
16
muscarinic acetylcholine receptors. Neurobiol Dis. 82:254-2(1.
17
39. Gard PR, Mandy A, Sutcliffe MA (1999–: Evidence of a possible role of altered angiotensin
18
function in the treatment, but not etiology, of depression. Biol Psychiatry. 45: 1030-4.
19
40. Costa-Ferreira M, Morais-Silva G, Gomes-de-Souza L, Marin MT, Crestani CC (2019–:
27
Luo H et al
1
The AT1 Receptor Antagonist Losartan Does Not Affect Depressive-Like State and Memory
2
Impairment Evoked by Chronic Stressors in Rats. Front Pharmacol. 10.
3
41. Tan J, Wang JM, Leenen FH (2005–: Inhibition of brain angiotensin-converting enzyme by
4
peripheral administration of trandolapril versus lisinopril in Wistar rats. Am J Hypertens.
5
18:158-1(4.
6
42. Okada Y, Tuchiya Y, Yagyu M, Kozawa S, Kariya K (1977–: Synthesis of bradykinin
7
fragments and their effect on pentobarbital sleeping time in mouse. Neuropharmacology.
8
1(:381-383.
9
43. Dalmolin GD, Silva CR, Belle NAV, Rubin MA, Mello CF, Calixto JB, et al. (2007–:
10
Bradykinin into amygdala induces thermal hyperalgesia in rats. Neuropeptides. 41:2(3-270.
11
44. Viel TA, Lima Caetano A, Nasello AG, Lancelotti CL, Nunes VA, Araujo MS, et al. (2008–:
12
Increases of kinin B1 and B2 receptors binding sites after brain infusion of amyloid-beta 1-40
13
peptide in rats. Neurobiol Aging. 29:1805-1814.
14
45. Bouhadfane M, Kaszas A, Rozsa B, Harris-Warrick RM, Vinay L, Brocard F (2015–:
15
Sensitization of neonatal rat lumbar motoneuron by the inflammatory pain mediator bradykinin.
16
Elife. 4:e0(195.
17
4(. Kozma R, Ahmed S, Best A, Lim L (1995–: The Ras-related protein Cdc42Hs and
18
bradykinin promote formation of peripheral actin microspikes and filopodia in Swiss 3T3
19
fibroblasts. Mol Cell Biol. 15:1942-1952.
28
Luo H et al
1
47. Sakurada K, Kato H, Nagumo H, Hiraoka H, Furuya K, Ikuhara T, et al. (2002–: Synapsin I
2
is phosphorylated at Ser(03 by p21-activated kinases (PAKs– in vitro and in PC12 cells
3
stimulated with bradykinin. The Journal of biological chemistry. 277:45473-45479.
4
48. Shen W, Wu B, Zhang Z, Dou Y, Rao ZR, Chen YR, et al. (200(–: Activity-induced rapid
5
synaptic maturation mediated by presynaptic cdc42 signaling. Neuron. 50:401-414.
6
49. Choi J, Ko J, Racz B, Burette A, Lee JR, Kim S, et al. (2005–: Regulation of dendritic spine
7
morphogenesis by insulin receptor substrate 53, a downstream effector of Rac1 and Cdc42
8
small GTPases. J Neurosci. 25:8(9-879.
9
50. Fang Y, Park IH, Wu AL, Du G, Huang P, Frohman MA, et al. (2003–: PLD1 regulates
10
mTOR signaling and mediates Cdc42 activation of S(K1. Curr Biol. 13:2037-2044.
11
51. Endo M, Antonyak MA, Cerione RA (2009–: Cdc42-mTOR signaling pathway controls
12
Hes5 and Pax( expression in retinoic acid-dependent neural differentiation. J Biol Chem.
13
284:5107-5118.
14
52. Radley JJ, Anderson RM, Hamilton BA, Alcock JA, Romig-Martin SA (2013–: Chronic
15
stress-induced alterations of dendritic spine subtypes predict functional decrements in an
16
hypothalamo-pituitary-adrenal-inhibitory prefrontal circuit. J Neurosci. 33:14379-14391.
17
53. Siltari A, Korpela R, Vapaatalo H (201(–: Bradykinin -induced vasodilatation: Role of age,
18
ACE1-inhibitory peptide, mas- and bradykinin receptors. Peptides. 85:4(-55.
19
54. Caetano AL, Dong-Creste KE, Amaral FA, Monteiro-Silva KC, Pesquero JB, Araujo MS,
29
Luo H et al
1
et al. (2015–: Kinin B2 receptor can play a neuroprotective role in Alzheimer's disease.
2
Neuropeptides. 53:51-(2.
3
55. Zhou JR, Shirasaki T, Soeda F, Takahama K (2014–: Cholinergic EPSCs and their
4
potentiation by bradykinin in single paratracheal ganglion neurons attached with presynaptic
5
boutons. J Neurophysiol. 112:933-941.
6
5(. Hedrick NG, Harward SC, Hall CE, Murakoshi H, McNamara JO, Yasuda R (201(–: Rho
7
GTPase complementation underlies BDNF-dependent homo- and heterosynaptic plasticity.
8
Nature. 538:104-108.
9
57. Kang HJ, Voleti B, Hajszan T, Rajkowska G, Stockmeier CA, Licznerski P, et al. (2012–:
10
Decreased expression of synapse-related genes and loss of synapses in major depressive
11
disorder. Nat Med. 18:1413-1417.
12
58.
13
renin-angiotensin pathway in posttraumatic stress disorder: angiotensin-converting enzyme
14
inhibitors and angiotensin receptor blockers are associated with fewer traumatic stress
15
symptoms. J Clin Psychiatry 73:849-55.
Khoury NM, Marvar PJ, Gillespie CF, Wingo A, Schwartz A, Bradley B, et al. (2012– The
16
17
18
Figure Legends
19
30
Luo H et al
1
Fig. 1 Captopril rapidly ameliorates CUS and CSDS-induced depressive-type
2
behaviors in mice. (A-B) Single intraperitoneal injection of captopril (9.75, 19.5 or
3
39 mg/kg) significantly reduced the immobility time in the TST (A) and FST (B) (n =
4
6-20, **p<0.01). (C) Time-concentration curve of captopril in the plasma, mPFC and
5
cerebrospinal fluid (CSF) of mice (19.5 mg/kg, i.p., 30 min: n=7-11, 1 h: n=7-11, 3 h:
6
n=6-8, 6 h: n=5-7, 12 h: n=3-8). (D) The effect of captopril (9.75, 19.5 or 39 mg/kg,
7
i.p) on sucrose preference and immobility time in TST and FST of CUS-treated mice
8
(n=7-16, *p<0.05, **p<0.01). (E) Sucrose preference was measured in the
9
CUS-treated mice at 1 d (n=6-11), 3 d (n=16-24) and 7 d (n=17-24) after captopril
10
injection (19.5 mg/kg, i.p., *p<0.05, **p<0.01 vs CON, ##p<0.01 vs CUS). (F) Social
11
index in the interaction zone of CSDS-treated mice was measured in social interaction
12
test 24 h after captopril administration (19.5 mg/kg, i.p., n=8-13, **p<0.01). Data are
13
expressed as mean ± S.E.M.
14
15
Fig. 2 Pharmacological regulators of angiotensin, such as DRI, ARB and Ang II
16
nAb, can not mimic the rapid antidepressant activity of ACEI. (A) Schematic
17
showing pharmacology of DRI, ACEI, ARB and Ang II nAb. (B) Aliskiren (10 and
18
50 mg/kg, i.p.) did not display rapid antidepressant activity in CUS-treated mice
19
(n=9-13). (C) Valsartan (30 or 60 mg/kg, i.p.) did not mimic the rapid antidepressant
20
activity of captopril in the CUS-treated mice (n=9-16). (D) Direct down-regulation of
21
central Ang II levels by locally infusion with Ang II nAb (1 µg/µl per side) did not 31
Luo H et al
1
mimic the rapid antidepressant activity of captopril (n=7-9). (E) Lisinopril (39.6
2
mg/kg, at the same molar equivalent dose of 19.5 mg/kg captopril), a long-lasting
3
ACEI, exerted similar rapid antidepressant activities to captopril in the FST (n=8,
4
**p<0.01 vs vehicle). (F) Lisinopril (39.6 mg/kg) exerted a faster and longer
5
antidepressant effect than that of captopril in the CUS-treated mice. Sucrose
6
preference at 2 h, 24 h, 7 d after systemic injection of lisinopril (39.6 mg/kg) and
7
captopril (19.5 mg/kg) in the CUS-treated mice (n=7-16, **p<0.01 vs CON, ##p<0.01
8
vs CUS). Data are expressed as mean ± S.E.M.
9
10
Fig. 3 Deficits in the BK signaling contributes to the CUS-induced
11
depressive-like behaviors in mice. (A) CUS did not affect the central expression of
12
RAS system. The level of Ang II, ACE, AT1R and AT2R were detected in the mPFC,
13
hippocampus, and NAc of the stressed mice (n=4-11). (B-D) The BK level
14
significantly decreased in the mPFC, which was reversed by captopril (B-C, n=9-13),
15
and in the plasma of CUS mice, which was also reversed by captopril (C-D). (E) BK
16
levels in the plasma of depressive patients were lower than those of healthy subjects
17
(n=7-12). (F) The expression of B1R and B2R in the mPFC of CUS mice (n=7-9). (G)
18
Infusion of BK (50 ng/µl per side) into the mPFC rapidly reversed CUS-induced
19
depressive-like behaviors in the SPT and TST (n=10-13). (H) Successive
20
administration of HOE140 (65.226 µg/kg per day, i.p, 7 days), but not DALBK
21
(99.818 µg/kg per day, i.p, 7 days) significantly increased the immobility time in the 32
Luo H et al
1
FST (n=10). Data are expressed as mean ± S.E.M. *p<0.05, **p<0.01.
2
3
Fig. 4 Both pharmacological and genetic blockade of B2R abolish the
4
antidepressant activity of captopril. (A) Locally bilateral infusion of HOE140, not
5
DALBK, abolished captopril’s effect on despair behavior in the TST and FST
6
(n=8-11). (B) HOE140 abolished captopril’s effect on the CUS-induced
7
depressive-like behaviors (n=12-15) in the SPT, TST and FST. (C) The stressed mice
8
were stereotaxically injected with GFP-tagging LV-B2R-shRNA or scrambled
9
shRNA. B2R knockdown abolished captopril’s effect on the CUS-induced
10
depressive-like behaviors in the SPT, TST and FST (n=12-15). Data are expressed as
11
mean ± S.E.M. *p<0.05, **p < 0.01.
12
13
Fig. 5 BK-stimulated Cdc42 activity confers the antidepressant effects of
14
captopril via activating the mTORC1 pathway. (A) Captopril reversed
15
CUS-induced defect in Cdc42 activity, which was assayed by detecting the
16
GTP-bound Cdc42 (n=9-11). (B) Locally bilateral infusion of ML141 blocked the
17
effect of captopril on immobility in TST and FST (n=8-9). (C) Locally bilateral
18
infusion of ML141 blocked the effect of captopril on CUS-induced depressive-like
19
behaviors (n = 9-11). (D) Captopril activated the mTORC1 pathway and pre-infusion
20
of ML141 into the mPFC blocked captopril’s effect on p-mTOR (n=4). **p<0.01. (E) 33
Luo H et al
1
Captopril (19.5 mg/kg, i.p) reversed the CUS-induced decrease in p-mTOR,
2
p-p70S6K and BDNF levels in the mPFC, which was abolished by LV-B2R-shRNA
3
(1 µl per side, n=4-7). (F) Pre-injection of rapamycin into mPFC blocked the effect of
4
captopril (19.5 mg/kg) on SPT and FST in CUS mice (n=7-11). Data are expressed as
5
mean ± S.E.M. *p<0.05, **p<0.01.
6
7
Fig. 6 Captopril reverses the CUS-induced synaptic loss and stimulates
8
synaptogenesis. (A) Mice were bilaterally injected with vehicle or ML141 into the
9
mPFC. After 30 min, mice were intraperitoneally injected with vehicle or captopril,
10
and the spine density was observed after 24 h using a confocal microscope.
11
Representative 3D reconstructing image of dendritic spines (A) and quantification of
12
average dendritic spine density (B, n=6-12) were shown. Scale bar: 5 µm. (C-D)
13
Captopril restored the CUS-induced decrease in synaptic proteins, including GluA1
14
and PSD95 in the mPFC (C, n=4) and hippocampus (D, n=4). (E) A pharmacology
15
model for the fast-acting antidepressant activity of ACEIs. ACEIs enter the central
16
nervous system, potentially inhibit the ACE activity and increase central BK level,
17
following by an activation of B2R-Cdc42-mTORC1-dependent synaptogenesis. Data
18
are expressed as mean ± S.E.M. *p<0.05, **p<0.01.
19
34