FCPR16, a novel phosphodiesterase 4 inhibitor, produces an antidepressant-like effect in mice exposed to chronic unpredictable mild stress

FCPR16, a novel phosphodiesterase 4 inhibitor, produces an antidepressant-like effect in mice exposed to chronic unpredictable mild stress

Accepted Manuscript FCPR16, a novel phosphodiesterase 4 inhibitor, produces an antidepressant-like effect in mice exposed to chronic unpredictable mil...

25MB Sizes 0 Downloads 58 Views

Accepted Manuscript FCPR16, a novel phosphodiesterase 4 inhibitor, produces an antidepressant-like effect in mice exposed to chronic unpredictable mild stress

Qiuping Zhong, Hui Yu, Chang Huang, Jiahong Zhong, Haitao Wang, Jiangping Xu, Yufang Cheng PII: DOI: Reference:

S0278-5846(18)30028-9 https://doi.org/10.1016/j.pnpbp.2018.10.017 PNP 9526

To appear in:

Progress in Neuropsychopharmacology & Biological Psychiatry

Received date: Revised date: Accepted date:

23 January 2018 28 September 2018 30 October 2018

Please cite this article as: Qiuping Zhong, Hui Yu, Chang Huang, Jiahong Zhong, Haitao Wang, Jiangping Xu, Yufang Cheng , FCPR16, a novel phosphodiesterase 4 inhibitor, produces an antidepressant-like effect in mice exposed to chronic unpredictable mild stress. Pnp (2018), https://doi.org/10.1016/j.pnpbp.2018.10.017

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. 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.

ACCEPTED MANUSCRIPT

FCPR16, a novel phosphodiesterase 4 inhibitor, produces an antidepressant-like effect in mice exposed to chronic unpredictable mild stress Qiuping Zhong 1, Hui Yu1, Chang Huang 1 , Jiahong Zhong 1, Haitao Wang 1, Jiangping Xu1,2 *, Yufang

PT

Cheng 1 *

RI

1, Neuropharmacology and Drug Discovery Group, School of Pharmaceutical Sciences, Southern Medical University,

SC

Guangzhou, China

NU

2, Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, China

MA

Abstract

The canonical phosphodiesterase 4 (PDE4) inhibitors produce antidepressant-like effects in a variety of animal models. However, severe side effects, particularly vomiting and nausea, limit their clinical

D

application. FCPR16 is a novel PDE4 inhibitor with less vomiting potential. However, whether it will

TE

exert an antidepressant-like effect remains unclear. Here, we aimed to evaluate the effect of FCPR16 in mice subjected to chronic unpredictable mild stress (CUMS). Our results showed that FCPR16 produced

EP

antidepressant-like effects in multiple behavioral tests, including a forced swimming test, tail suspension

AC C

test, sucrose preference test and novelty suppression feeding test. Simultaneously, data indicated that FCPR16 enhanced the levels of several proteins, including cAMP, brain derived neurotrophic factor, exchange protein directly activated by cAMP 2 (EPAC-2), synapsin1, postsynaptic density protein 95, phosphorylated cAMP response element binding protein and extracellular regulated protein kinases 1/2, which were downregulated by CUMS in both the cerebral cortex and hippocampus. The number of DCX+ cells in the hippocampus of CUMS mice was increased after FCPR16 treatment. Moreover, treatment with FCPR16 resulted in decreased expression of pro-inflammatory cytokines (TNF-α, IL-6, and IL-1β)

ACCEPTED MANUSCRIPT

and increased expression of anti-inflammatory cytokines (IL-10) in mice challenged with CUMS. Consistently, the mRNA levels of microglial M1 markers (iNOS and TNF-α) were downregulated, while

PT

M2 markers (Arginase 1 and CD206) were upregulated in CUMS-exposed mice after FCPR16 treatment. Immunofluorescence analysis showed that FCPR16 inhibited the activation of microglial cells and

RI

increased the number of CD206+ in CUMS-exposed mice. Collectively, these results suggested that FCPR16 is a potential compound with effects against depressive-like behaviors, and the

SC

antidepressant-like effect of FCPR16 is possibly mediated through activation of the cAMP -mediated signaling pathways and inhibition of neuroinflammation in both the cerebral cortex and hippocampus.

NU

Keywords

AC C

EP

TE

D

MA

Depression, phosphodiesterase 4 inhibitors, FCPR16, neuroinflammation, microglial phenotypes

ACCEPTED MANUSCRIPT

1. Introduction Depression is a highly prevalent mental disorder. According to a report from the World Health

PT

Organization (WHO, 2017), over 300 million individuals are suffering from depression. The prevalence rate has been increasing year by year over the past decade. Approximately 800 thousand people die from

RI

suicide every year. Depression has become a heavy burden on society and the economy. However, the defects of clinical drugs increase the difficulty for antidepressant therapy. The effects of existing drugs

SC

usually emerge after 6-8 weeks treatment (Uher et al., 2011). Only less than 50% of patients effectively respond to drugs that are currently on the market (Geddes and Miklowitz, 2013). The rate of adverse

NU

effects is high (39% suicide, 60% feeling emotionally numb or others) (Read et al., 2014). The selective serotonin reuptake inhibitors (SSRIs), which are commonly used medications for depression treatment,

MA

have been reported to accelerate the transition from an insulin-resistant state to overt diabetes (Isaac et al., 2013). Searching for new therapeutic targets is particularly urgent.

D

Phosphodiesterase 4 (PDE4), an enzyme that selectively hydrolyzes cAMP, is highly expressed in neurons

TE

and glial cells (Lakics et al., 2010). PDE4 inhibitors exhibit antidepressant-like effects by increasing intracellular cAMP (Garcia et al., 2016). Elevated cAMP triggers a series of signaling cascades, which

EP

ultimately regulate a variety of pathophysiological processes. The cAMP/protein kinase A (PKA)/cAMP response element binding protein (CREB) signaling pathway is an important mediator in the regulation of

AC C

central nervous system (CNS) functions, including neuronal survival, axonal regeneration and synaptic plasticity (Conti and Beavo, 2007; Plattner et al., 2015). Deficits in these processes are closely linked to the development of depression. Exchange proteins directly activated by cAMP (EPACs), a family of cAMP sensor proteins that include isoforms EPAC-1 and EPAC-2, were first reported in 1998 (Kawasaki et al., 1998). EPAC-2 is highly expressed in the CNS (Wang et al., 2017) and has been implicated in the regulation of neurotransmitter release, neuronal differentiation, neurite growth, memory, learning, and a variety of brain diseases (Schmidt et al., 2013). An in vivo study (Liebenberg et al., 2011) showed that a

ACCEPTED MANUSCRIPT

cAMP analogue that inhibits PKA but not EPAC (Rp-8-Br-cAMP) still produced antidepressant-like effects in rats during the forced swimming test (FST) and increased the phosphorylation of CREB. The

PT

results implied that in addition to activating the cAMP/PKA/CREB signaling pathway, elevated cAMP may also stimulate EPAC and upregulate the proteins downstream of EPAC to mediate antidepressant

RI

activity. On the other hand, an abundant number of studies have shown that long-term stress is accompanied by neuroinflammation (Howren et al., 2009). Elevated inflammatory cytokines are also a

SC

critical pathogeny of depression (Kubera et al., 2011), and downregulation of inflammatory levels could exert neuroprotective functions and reverse depressive-like behaviors (Kohler et al., 2014). A previous

NU

study has shown that the PDE4 inhibitor rolipram reversed amyloid β-induced memory deficits in rats via suppression of neuronal inflammation (Wang et al., 2012). Neuroinflammation suppression is one of the

MA

ways that PDE4 inhibitors exert antidepressant-like effects. Basic and clinical studies (Price and Drevets, 2010; MacQueen and Frodl, 2011) demonstrated that depression was associated with a reduced size of

D

and fewer synapses in the cerebral cortex and hippocampus, the brain regions for regulating mood. It was

TE

accompanied by a decline in the expression of synapse-related protein, i.e., synapsin 1 and postsynaptic density protein 95 (PSD95). Brain derived neurotropic factor (BDNF), a protein downstream of the

EP

cAMP/PKA/CREB signaling pathway, promotes neuronal development early in life and maintains synaptic plasticity in the adult brain (Krishnan and Nestler, 2008). PDE4 inhibitors can reverse synaptic

AC C

dysfunction through activation of the cAMP/PKA/CREB signaling pathway. However, rolipram, a first generation PDE4 inhibitor, displayed a strong antidepressant-like effect in preclinical studies as well as in phase II clinical trials (Scott et al., 1991) but was not approved for clinical use

due

to

the

severe

emesis

effect.

FCPR16

(N-(2-chlorophenyl)-3-cyclopropylmethoxy-4-difluoromethoxybenzamide), a structural analogue of the traditional PDE4 inhibitor with 90 nM affinity for PDE4 (Zhou et al., 2016), which was designed and synthesized in our laboratory, did not cause emesis during the 180 min observation period at 3 mg/kg in

ACCEPTED MANUSCRIPT

Beagle dogs (Chen et al., 2017). Previous studies (Zhou and Ge et al., 2016) showed that FCPR16 reduced the expression of tumor necrosis factor- (TNF-α) in lipopolysaccharide (LPS)-exposed BV-2

PT

cells and improved the survival rate of rats in a middle cerebral artery occlusion (MCAO) model by upregulating cAMP-mediated signaling pathways and suppressing the expression of inflammation

RI

cytokines (Chen and Yu et al., 2017). Nevertheless, whether FCPR16 has an antidepressant potency is unclear. In the current study, we evaluated the antidepressant-like effect of FCPR16 via behavior tests in a

SC

mouse model of chronic unpredictable mild stress (CUMS) and investigated the mechanism. We hypothesized that FCPR16 would exert an antidepressant-like effect in CUMS-exposed mice by

NU

regulating the cAMP/PKA/CREB/BDNF and cAMP/EPAC/extracellular signal-regulated kinase (ERK) signal pathways and exerting anti-neuroinflammation. Our results showed that FCPR16 improved the

MA

depressant-like behaviors of CUMS mice in FST, tail suspension test (TST), sucrose preference test (SPT) and novelty-suppressed feeding test (NSFT), which was implicated in the upregulation of cAMP, BDNF,

D

EPAC-2, synapsin 1 and PSD95 levels as well as the phosphorylation of CREB. FCPR16 treatment

TE

downregulated the expression of the inflammatory cytokines TNF-, interleukin-6 (IL-6) and interleukin-1 (IL-1) and promoted shifting of the microglial phenotype from M1 to M2 in the cerebral

EP

cortex and hippocampus.

AC C

2. Materials and methods 2.1. Animals

Male C57BL/6 mice (18-22 g) were purchased from the Laboratory Animal Centre of Southern Medical University (Guangzhou, China) and allowed to acclimate for 1 week before the experiments. The mice were housed individually under standard conditions (22 ± 2ºC, 50%-70% humidity, 12-12 h light/dark cycle with lights on at 07:00) with access to food and water ad libitum. All experimental procedures were carried out in accordance with the NIH Guide for the Care and Use of Laboratory Animals (NIH, Eighth

ACCEPTED MANUSCRIPT

Edition, 2011) and approved by the Laboratory Animal Ethics Committee of Southern Medical University. Studies from animal experiments showed that behaviors and monoamine transmitter releases in rodents

PT

could be influenced by estrogen fluctuations (Fink et al., 1996; Okada et al., 1997; Galea et al., 2001; Dalla et al., 2004). To avoid the impact of estrogen on depressive-like behaviors in our experiments, male

RI

mice were selected for this study. 2.2. Compound

SC

Escitalopram was purchased from Aladdin (Shanghai, China). FCPR16 (Figure 1(a)) was structurally reconstructed and synthesized by Professor Zhou (Zhou and Ge et al., 2016). These compounds were in

0.5%

sodium

carboxymethylcellulose.

NU

suspended

carboxymethylcellulose.

vehicle

was

0.5%

sodium

MA

2.3. Experimental designs

The

The experiment included two parts. Part I evaluated the antidepressant-like potency of FCPR16 in the

D

behavioral despair model (Wang et al., 2017). Mice were randomly divided into 6 groups (n=8-12 per

TE

group) using the random number program in SPSS software and received vehicle (0.1 ml/10 g), positive control escitalopram (10 mg/kg, i.g.) or FCPR16 (0.35 mg/kg, 0.7 mg/kg, 1.4 mg/kg and 2.8 mg/kg, i.g.).

EP

Forth-five minutes after a single compound administration, mice were subjected to forced swimming test (FST), open field test (OFT) or TST. OFT was taken as an evaluation of locomotor activity. Part II further

AC C

confirmed the antidepressant-like effect in the CUMS mice and explored the mechanism of the effect. Mice were randomly divided into 4 groups including one control and three experimental groups (n=12-14 per group) using the random number program in SPSS software. Experimental groups were subjected to CUMS as described below for 6 weeks. After CUMS, the experimental groups received FCPR16 (1.5 mg/kg, i.g.), positive control escitalopram (10 mg/kg, i.g.), or vehicle once daily for 3 weeks (0.1 ml/10 g). The control group was not exposed to stressors and received vehicle. The FST and TST were conducted at the baseline (6 weeks after CUMS and prior to drug treatment) and upon completion of the

ACCEPTED MANUSCRIPT

3-week drug treatment. The SPT and NSFT were conducted after treatment. After the behavior tests were completed, biochemical tests were conducted (Figure 2).

PT

2.4. Chronic unpredictable mild stress (CUMS) The CUMS procedure was performed as described previously (Willner, 1997; Thakare et al., 2017) with

RI

minor modifications. It contained 7 different stressors (Table 1) randomly conducted across 9 weeks. The same stressor was not used on two consecutive days.

SC

Table 1. Stressors used for CUMS. Stressor

1

restriction of behaviors

4h

2

water and food deprivation

18 h

3

45º cage tilt

18 h

4

swimming in water at 4-6ºC

5 min

5

tail pinch

5 min

6

water deprivation

MA

D

TE

illumination

Duration

24 h overnight

EP

7 2.5. Behavior test

NU

Number

2.5.1. Sucrose preference test (SPT)

AC C

The SPT was performed as described previously (Willner et al., 1987; Zhao et al., 2014) with slight modifications. Mice were individually placed in cages with two bottles. On day 1, two bottles contained water. On day 2, one bottle contained water and the other contained 1% sucrose. Two of the bottles’ positions were switched and kept until 12 h later. On day 3, mice were deprived of water and food for 24 h. On day 4, the SPT was conducted and the test session was 4 h. Two bottles contained water and 1% sucrose and the positions were switched 2 h later. The sucrose preference ratio (SPR) was calculated as

ACCEPTED MANUSCRIPT

follows: [sucrose intake (g)/ (sucrose intake (g) + water (g))] × 100%. 2.5.2. Forced swimming test (FST) Mice were forced to swim individually in a cylindrical glass container (diameter: 13 cm, height: 24 cm,

PT

depth: 14 cm) (Castagne et al., 2011) that contained water (22  2ºC). Each session lasted for 6 min and

RI

was recorded with a video camera. The immobility time was scored for the last 4 min of the session. 2.5.3. Tail suspension test (TST)

SC

The TST was conducted as described previously (Castagne and Moser et al., 2011). Mice were suspended, and the experimenter made sure that the mice hung with their tails in a straight line. The testing session

NU

lasted for 6 min, and the immobility time was recorded. 2.5.4. Open-field test (OFT)

MA

OFT was conducted with minor modifications (Cui et al., 2012). Mice were placed individually in the center of a box (60 cm × 60 cm × 20 cm) divided into 4 quadrants of equal areas with 36 lattices and

D

recorded for 6 min. The numbers of rearing (the mice stood on rear limbs) and crossing (the mice crossed

TE

the lattices with four paws) were scored during the last 5 min. 2.5.5. Novelty-suppressed feeding test (NSFT)

EP

The NSFT was conducted as described previously (Li et al., 2017). Mice were deprived of food for 24 h and placed in a box (60 cm × 60 cm × 20 cm) with fresh bedding. Food was put in the center of the box,

AC C

and mice were placed in a corner of the box. The latency of first-time feeding was recorded. 2.6. Western blot

Western blot analyses were performed as previously described (Zou et al., 2017). Briefly, proteins were extracted using 1×RIPA lysis buffer containing 1% protease inhibitor cocktail and 1% phosphatase inhibitor cocktail (Sigma, USA). The protein concentration was measured using the BCA method (Thermo Scientific, USA). Proteins were separated by SDS-PAGE, electrotransferred, blocked and then incubated a primary antibody overnight at 4ºC. The following primary antibodies were used: GAPDH

ACCEPTED MANUSCRIPT

(ab181602, abcam, UK), ERK1/2 (WL01864, Wanleibio, China), p-ERK1/2 (WLP003, Wanleibio), EPAC-2 (ab193665, abcam), CREB (#9197, CST, USA), p-CREB (#9198, CST), BDNF (SAB2108004,

PT

Sigma), synapsin 1 (ab64581, abcam), and PSD95 (ab18258, abcam). The blots were then incubated with a secondary antibody for 2 h at room temperature before visualization with an ECL method and analysis

RI

by ImageJ software. GAPDH was used as an internal control. 2.7. Enzyme-linked immunosorbent assay (ELISA)

SC

The cerebral cortex and hippocampus of mice were quickly dissected on ice, homogenized and centrifuged at 12000 × rpm for 15 min at 4ºC. Supernatants were used to determine the concentrations of

NU

IL-6 (EK0411, BOSTER, China), TNF-α (EK0527, BOSTER), IL-10 (EK0417, BOSTER), IL-1β (EK0394, BOSTER) and cAMP (KGE012B, R&D system, USA) by ELISA kits. Data were all

MA

normalized to total protein.

2.8. RNA extraction and real-time PCR (RT-PCR)

D

Total RNA was extracted using Trizol reagent (9109, TaKaRa, Japan) and reverse transcribed (RR036A)

TE

and amplified (RR420A) using a kit from TaKaRa. The RT-PCR conditions were as follows: initial 95ºC for 30 s follow by 40 cycles at 95ºC for 5 s and extension at 60ºC for 31 s. Data were normalized against

EP

GAPDH and are presented as fold changes. Primer sequences are listed in Table 2. Table 2. Primer sequences used in RT-PCR experiments. Reverse (5’ to 3’)

AGGTCGGTGTGAAACGGATTTG

TGTAGACCATGTAGTTGAGGTCA

GTTCTCAGCCCAACAATACAAGA

GTGGACGGGTCGATGTCAC

CCCTCACACTCAGATCATCTTCT

GCTACGACGTGGGCTACAG

Arg1

CTCCAAGCCAAAGTCCTTAGAG

AGGAGCTGTCATTAGGGACATC

CD206

CTCTGTTCAGCTATTGGACGC

CGGAATTTCTGGGATTCAGCTTC

GAPDH iNOS TNF-α

AC C

Forward (5’ to 3’)

Gene

Arg1: Arginase 1

ACCEPTED MANUSCRIPT

2.9. Immunohistochemistry Mice were perfused with ice-cold phosphate-buffered saline and 4% paraformaldehyde. Brains were

PT

postfixed in 4% paraformaldehyde and dehydrated in graded sucrose solutions. Brains sections were prepared using a freezing microtome (Leica, Germany). Sections were washed and permeated by 0.3%

RI

Triton for 15 min. After blocking at room temperature for 1 h, sections were incubated with rat anti-CD206 (GTX42264, GeneTex, USA), rabbit anti-Iba1 (019-19741, Wako, Japan) or guinea pig

SC

anti-doublecortin (ab2253, Millipore, USA) overnight at 4ºC. Subsequently, sections were incubated with an appropriate secondary antibody at room temperature for 2 h and DAPI for 10 min. The secondary

NU

antibodies included Alexa Fluor® 555 goat anti-rat IgG (405420, Biolegend, USA), Dylight 488 goat anti-rat IgG (A23220, Abbkine, USA) and Alexa Fluor 488 AffiniPure goat anti-guinea pig IgG (H+L)

MA

(106-545-003, Jackson, USA). Image analysis was performed by a confocal microscope (Nikon, Japan). For quantification of immunofluorescence, 6 fields from each group were analyzed by Nikon Imaging

D

Elements Software. For each field, the total number of Iba1-positive cells and CD206-positive cells were

2.10. Statistical analysis

TE

counted.

EP

All data are presented as the mean ± S.E.M. All statistical comparisons were computed using SPSS 19.0 software. When normality and equal variance were achieved, potential differences between the mean

AC C

values were analyzed with a one-way analysis of variance (ANOVA) followed by Bonferroni’s post hoc test. In CUMS model, at week 6, TST and FST data between the control and CUMS group were analyzed with one-way ANOVA. At week 9, data of TST and FST were analyzed with repeated ANOVA(week as a within factor (repeated factor)) (Li et al., 2009). For evaluation of the NSFT results, Kaplan-Meier survival analysis was used followed by the Mantel-Cox log-rank test. P-value of less than 0.05 was reported as statistically significant. All experiments were conducted in a blinded manner, experimenters did not know the treatment conditions.

ACCEPTED MANUSCRIPT

3. Results 3.1. FCPR16 ameliorated depressant-like behaviors in mice

PT

To evaluate whether the compound FCPR16 has an antidepressant-like effect, we conducted FST and TST 45 min after a single administration. In the behavioral despair model, compared with the control group,

RI

FCPR16 (1.4 mg/kg and 2.8 mg/kg) and the positive control escitalopram both decreased the immobility time in the FST (F (5, 60) = 6.998, p < 0.001, Figure 1(b)) and the TST (F (5, 60) = 9.521, p < 0.001,

SC

Figure 1(c)). Neither agent altered the number of crossing (F (5, 60) = 1.105, p > 0.05, Figure 1(d)) or rearing (F (5, 60) = 2.008, p > 0.05, Figure 1(e)) in the OFT. These findings suggested that the effect of

NU

FCPR16 to decrease the immobility time in FST and TST was not a result of psychostimulant disturbances. FCPR16 may have the potential to improve depressant-like behaviors in mice.

MA

To further investigate the antidepressant-like effect of FCPR16, mice were exposed to CUMS for 6 weeks and assessed with the TST, FST, SPT and NSFT after treatment for another 3 weeks (Figure 2). At week 6,

D

in the FST (F (3, 50) = 8.362, p < 0.001, Figure 3(a)) and TST (F (3, 48) = 6.775, p < 0.001, Figure 3(c)),

TE

the immobility time in CUMS-exposed groups showed significant increases compared with that in the control group without CUMS. A one-way ANOVA revealed that there was no significant difference (p >

EP

0.05) between the CUMS (CUMS + vehicle), FCPR16 (CUMS + FCPR16) and escitalopram (CUMS + Escitalopram) groups on immobility times before treatments.

AC C

At week 9, for the FST, a repeated ANOVA showed significant effects for week (F (1, 50) = 15.613, p < 0.001), group (F (3, 50) = 13.239, p < 0.001) and group × week interaction (F (3, 50) = 3.966, p < 0.05) on immobility time (Figure 3(b)). A post hoc test showed that immobility time was significantly decreased after FCPR16 (p < 0.01) or escitalopram (p < 0.05) treatment compared with the CUMS group. In the TST, a repeated ANOVA showed significant effects for week (F (1, 48) = 23.605, p < 0.001), group (F (3, 48) = 7.213, p < 0.001) and group × week interaction (F (3, 48) = 3.989, p < 0.05) on immobility time (Figure 3(d)). Post hoc test results showed that, compared with the CUMS group, the immobility

ACCEPTED MANUSCRIPT

times for mice were significantly decreased after FCPR16 (p < 0.05) or escitalopram (p < 0.05) treatment. In the SPT, the SPR of CUMS-exposed mice showed a significant decrease compared with the control

PT

group. However, FCPR16 (p < 0.05) and escitalopram (p < 0.05) treatment attenuated the effect of CUMS (F (3, 50) = 4.295, p < 0.01, Figure 3(e)) and increased the SPR of mice after 3 weeks treatment

RI

compared with the CUMS group. In NSFT, the mean latency in CUMS-exposed mice (166.3 s) was significantly longer than that of the controls (96.9 s) as well as the FCPR16 (108.7 s) and escitalopram

SC

(120.9 s) treatment groups (Figure 3(f)). In summary, FCPR16 produced an antidepressant-like effect in

AC C

EP

TE

D

MA

NU

CUMS-exposed mice.

AC C

EP

TE

D

MA

NU

SC

RI

PT

ACCEPTED MANUSCRIPT

Figure 1. Effect of FCPR16 in mice in the behavioral despair model. Chemical structure of FCPR16 (N-(2-chlorophenyl)-3-cyclopropylmethoxy-4-difluoromethoxybenzamide) (a). Immobility time in the FST (b) and TST (c). The number of crossing (d) and rearing (e) in the OFT. Data are shown as the mean ± S.E.M. *P<0.05, ***P<0.001 vs the control group. n = 8-12.

MA

NU

SC

RI

PT

ACCEPTED MANUSCRIPT

D

Figure 2. The experimental schematic. Mice were divided into 4 groups: one control group and three

TE

experimental groups. Experimental groups were subjected to CUMS as described below for 6 weeks. TST and FST were conducted at week 6 to demonstrate the successful construction of the depression model in

EP

mice. Then, mice in the experimental groups received FCPR16, positive control escitalopram and vehicle once daily for 3 weeks. The control group also received the same volume vehicle once daily for 3 weeks. 3

weeks

treatment,

AC C

After

the

mice

performed

the

TST,

FST,

SPT

and

NSFT.

AC C

EP

TE

D

MA

NU

SC

RI

PT

ACCEPTED MANUSCRIPT

Figure 3. Antidepressant-like effect of FCPR16 in CUMS-exposed mice. The immobility time of mice in FST after 6 weeks CUMS and prior to treatment (a) and after 3 weeks treatment (b). The immobility

ACCEPTED MANUSCRIPT

time of mice in TST after 6 weeks CUMS and prior to treatment (c) and after 3 weeks treatment (d). The sucrose preference ratio in the SPT (e) and the result in the NSFT (f) after 3 weeks treatment. Data are

RI

CUMS group. n = 12-14. FCPR16: 1.5 mg/kg. Escitalopram: 10 mg/kg.

PT

shown as the mean ± S.E.M. *p<0.05, **p<0.01, *** p<0.001 vs the control group. #p<0.05, ##p<0.01 vs the

3.2. FCPR16 upregulated cAMP-mediated signaling pathways and synapse-related protein

SC

expression in the cerebral cortex

To evaluate whether FCPR16 has a regulatory role in the cAMP canonical pathways, cAMP/PKA/CREB

NU

and cAMP/EPAC/ERK expression levels were measured by Western blotting. In the cerebral cortex of mice, the results showed that FCPR16 treatment reversed the CUMS-induced reduction in cAMP levels

MA

(F (3, 8) = 15.900, p<0.01, Figure 4(a)) and upregulated the protein expression of cAMP-mediated signaling pathways, including phosphorylation of CREB (F (3, 8) = 12.130, p<0.01, Figures 4(b) and (e)),

D

BDNF (F (3, 8) = 15.740, p<0.01, Figures 4(b) and (f)) and EPAC-2 (F (3, 8) = 19.390, p<0.001, Figures

TE

4(c) and (g)). However, the phosphorylation of ERK1/2 (F (3, 12) = 2.634, p>0.05, Figures 4(c) and (h)) was increased slightly without statistical significance in both the FCPR16 and escitalopram groups

EP

compared with the CUMS group. The above data suggested that FCPR16 exerted an antidepressant-like effect accompanied by upregulation of the cAMP/CREB/BDNF and cAMP/EPAC-2 pathways.

AC C

To examine the protective effect of FCPR16 on synapse function in the cerebral cortex of CUMS-exposed mice, we measured the expression levels of synapse-associated proteins synapsin 1 and PSD95. The results showed decreased expression of synapsin 1 and PSD95 in the cerebral cortex of mice upon CUMS. However, FCPR16 treatment reversed the alteration and increased the expression levels of synapsin 1 (F (3, 8) = 19.330, p<0.001, Figure 4(d) and (i)) and PSD95 (F (3, 8) = 13.820, p<0.01, Figure 4(d) and (j)) compared with the CUMS group.

ACCEPTED MANUSCRIPT

3.3. FCPR16 upregulated cAMP-mediated signaling pathways and synapse-related protein expression in the hippocampus

PT

In addition to the cerebral cortex, the hippocampus is one of the brain regions associated with depression. Thus, we also measured the expression levels of the above proteins in the hippocampus. The results

RI

showed that the changes in protein expression levels in the hippocampus were similar to those of the cerebral cortex. On one hand, FCPR16 treatment upregulated the level of cAMP (F (3, 8) = 16.140,

SC

p<0.001, Figure 5(a)) and activated cAMP-mediated signaling pathways, increasing the protein expression of BDNF (F (3, 8) = 11.690, p<0.01, Figure 5(b) and (f)), EPAC-2 (F (3, 8) = 9.081, p<0.01,

NU

Figure 5(c) and (g)) and phosphorylated CREB (F (3, 8) = 6.057, p<0.05, Figure 5(b) and (e)) compared with the CUMS group. Similarly, there was no significant difference in the changes in phosphorylation of

MA

ERK1/2 (F (3, 12) = 3.138, p>0.05, Figure 5(c) and (h)) between the CUMS and FCPR16 group. On the other hand, FCPR16 treatment reversed CUMS-induced decreases in the expression of synapse-related

D

protein synapsin 1 (F (3, 12) = 11.920, p<0.001, Figure 5(d) and (i)) and PSD95 (F (3, 12) = 5.567,

AC C

EP

TE

p<0.05, Figure 5(d) and (j)).

AC C

EP

TE

D

MA

NU

SC

RI

PT

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIPT

Figure 4. Effect of FCPR16 on the regulation of signaling pathways in the cerebral cortex of mice. The levels of cAMP (a), p-CREB (b, e), BDNF (b, f), EPAC-2 (c, g) p-ERK1/2 (c, h), synapsin 1 (d, i) and PSD95 (d, j). Data are shown as the mean ± S.E.M. *p<0.05, **p<0.01, ***p<0.001 vs the control group. p<0.05,

##

p<0.01,

###

PT

#

p<0.001 vs the CUMS group. n = 3-4 samples per group (repeated 3 times per

AC C

EP

TE

D

MA

NU

SC

RI

sample).

AC C

EP

TE

D

MA

NU

SC

RI

PT

ACCEPTED MANUSCRIPT

Figure 5. Effect of FCPR16 on the regulation of signaling pathways in the hippocampus of mice. The level of cAMP (a), p-CREB (b, e), BDNF (b, f), EPAC-2 (c, g), p-ERK1/2 (c, h), synapsin 1 (d, i) and

ACCEPTED MANUSCRIPT

PSD95 (d, j). Data are shown as the mean ± S.E.M. *p<0.05, **p<0.01, ***p<0.001 vs the control group. #

p<0.05,

##

p<0.01,

###

p<0.001 vs the CUMS group. n = 3-4 samples per group (repeated 3 times per

PT

sample).

RI

3.4. FCPR16 promoted neurogenesis in the hippocampus of mice exposed to CUMS FCPR16 treatment upregulated the cAMP signaling pathways and the expression levels of

SC

synaptic-related proteins. To evaluate whether FCPR16 improves neurogenesis in the hippocampus of CUMS-treated mice, we measured the number of DCX+ cells in the hippocampus by immunofluorescence

NU

and detected the expression of DCX by Western blotting. The number of DCX+ cells in the hippocampus of mice, indicative of the numbers of neuronal progenitor cells, was increased after FCPR16 treatment

MA

compared with the CUMS group (Figure 6 (a)). The Western blot results suggested that the expression level of DCX was higher in the hippocampus of mice treated with FCPR16 (F (3, 12) = 16.75, p<0.001,

AC C

EP

TE

D

Figure 6 (b) and (c)) than that of the CUMS group.

TE

D

MA

NU

SC

RI

PT

ACCEPTED MANUSCRIPT

EP

Figure 6. Effect of FCPR16 on Doublecortin (DCX) in the hippocampus of mice. DCX (green) and DAPI

AC C

(blue) were measured by immunofluorescence (a). White arrows point to DCX+ cells. Scale bar = 100 μm. The expression of DCX was measured by Western blotting (b, c). Data are shown as the mean ± S.E.M. ***

p<0.001 vs the control group. #p<0.05, ##p<0.01 vs the CUMS group. n = 4 per group (repeated 3 times

per sample).

ACCEPTED MANUSCRIPT

3.5. FCPR16 suppressed inflammatory cytokine expression in the cerebral cortex of CUMS mice To examine whether FCPR16 regulates inflammatory cytokine expression, inflammatory cytokines were measured by ELISA kits. In the cerebral cortex, as shown in the results, CUMS increased the expression

PT

of the pro-inflammatory cytokines TNF-α, IL-6, and IL-1β compared with the control group, while it

RI

decreased the anti-inflammatory cytokine IL-10. With FCPR16 treatment, the expression levels of TNF-α (F (3, 16) = 8.711, p<0.01, Figure 7(a)), IL-6 (F (3, 16) = 4.137, p<0.05, Figure 7(b)) and IL-1β (F (3, 16)

SC

= 7.333, p<0.01, Figure 7(c)) were significantly lower than those of CUMS-exposed mice. The level of the anti-inflammatory cytokine IL-10 in FCPR16-treated mice was higher than that in mice exposed to

NU

CUMS (F (3, 16) = 2.733, p>0.05, Figure 7(d)), but there was no significant difference.

MA

3.6. FCPR16 suppressed inflammatory cytokine expression in the hippocampus of CUMS mice In the hippocampus, CUMS also upregulated the pro-inflammatory cytokines TNF-α, IL-6, and IL-1β,

D

while it downregulated the anti-inflammatory cytokine IL-10. However, after FCPR16 treatment, the

TE

levels of pro-inflammatory cytokines TNF-α (F (3, 16) = 4.005, p<0.05, Figure 7(e)), IL-6 (F (3, 16) = 20.200, p<0.001, Figure 7(f)) and IL-1β (F (3, 16) = 62.440, p<0.001, Figure 7(g)) were decreased, while

EP

the level of anti-inflammatory cytokine IL-10 (F (3, 16) = 37.900, p<0.001, Figure 7(h)) was increased compared with the CUMS group. Our results demonstrated that FCPR16 protected mice against

AC C

CUMS-induced neuroinflammation in both the cerebral cortex and hippocampus.

AC C

EP

TE

D

MA

NU

SC

RI

PT

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIPT

Figure 7. Effect of FCPR16 on the release of inflammatory cytokines. The expression of pro-inflammatory cytokines TNF-α (a, e) and IL-6 (b, f), IL-1β (c, g) and anti-inflammatory cytokine IL-10 (d, h) in the cerebral cortex and hippocampus, respectively. Data are shown as the mean ± S.E.M. p<0.05,

***

p<0.001 vs the control group. # p<0.05,

##

p<0.01,

###

PT

*

p<0.001 vs the CUMS group. n = 5

RI

samples per group (each sample was measured in triplicates).

SC

3.7. FCPR16 improved the shift in microglial phenotype from M1 to M2 in the cerebral cortex The above results suggested that FCPR16 suppressed the expression of pro-inflammatory cytokines in the

NU

cerebral cortex and hippocampus of CUMS-exposed mice. Thus, to further confirm whether FCPR16 had effects on regulating the microglial phenotypes, we examined the mRNA levels of microglial M1 (TNF-

MA

and iNOS) and M2 (Arg1 and CD206) markers by RT-PCR. In the cerebral cortex, the results showed that FCPR16 decreased the CUMS-induced increases in the mRNA levels of the M1 markers TNF-α (F (3, 12)

D

= 14.230, p<0.001, Figure 8(a)) and iNOS (F (3, 12) = 17.450, p<0.001, Figure 8(b)). The mRNA levels

TE

of the M2 markers Arg1 (F (3, 8) = 64.390, p<0.001, Figure 8(c)) and CD206 (F (3, 8) = 12.610, p<0.01, Figure 8(d)) in the FCPR16 group were higher than those in the CUMS group.

EP

To further verify the results, immunofluorescence double labeling was performed using microglial marker Iba1 and microglial M2 marker CD206 (Figure 9 (a)). The ratio of CD206 + cells to Iba1 + cells (F (3, 20) =

AC C

6.249, p<0.01, Figure 9(a) and (c)) was increased in the cerebral cortex of the FCPR16-treated group compared with the CUMS group.

3.8. FCPR16 improved the shift in microglial phenotype from M1 to M2 in the hippocampus In the hippocampus, FCPR16 decreased the mRNA levels of the M1 markers TNF- (F (3, 16) = 9.213, p<0.001, Figure 8(e)) and iNOS (F (3, 12) = 15.44, p<0.001, Figure 8(f)) and increased the mRNA levels of the M2 markers Arg1 (F (3, 8) = 4.724, p<0.05, Figure 8(g)) and CD206 (F (3, 8) = 32.600, p<0.001,

ACCEPTED MANUSCRIPT

Figure 8(h)) compared with those in the CUMS group. The results of immunofluorescence showed that the ratio of CD206 + cells to Iba1 + cells (F (3, 20) = 56.670, p<0.001, Figure 9(b) and (d)) was increased in

PT

mice treated with FCPR16 compared with the CUMS group. Taken together, FCPR16 produced an anti-neuroinflammation effect and promoted a shift in microglial

AC C

EP

TE

D

MA

NU

SC

RI

phenotype from M1 to M2.

AC C

EP

TE

D

MA

NU

SC

RI

PT

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIPT

Figure 8. Effects of FCPR16 on microglial polarization. The mRNA level of the microglial M1 markers TNF-α (a, e) and iNOS (b, f) and the microglial M2 markers Arginase 1 (Arg1) (c, g) and CD206 (d, h) in the cerebral cortex and hippocampus, respectively. Data are shown as the mean ± S.E.M. * p<0.05, p<0.01,

***

p<0.001 vs the control group. #p<0.05, ##p<0.01,

###

PT

**

p<0.001 vs the CUMS group. n = 3-5

AC C

EP

TE

D

MA

NU

SC

RI

samples per group (each sample was measured in triplicate).

AC C

EP

TE

D

MA

NU

SC

RI

PT

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIPT

Figure 9. Effect of FCPR16 on the microglial immunofluorescence marker (Iba1) and the microglial M2 marker (CD206) in the cerebral cortex (a) and hippocampus (b), and their quantification, respectively (c, d). Scale bar = 100 μm. White arrows point out Iba+ cells. Green arrows point out CD206 + cells. Data are

RI

PT

shown as the mean ± S.E.M. ##p<0.01, ###p<0.001 vs the CUMS group. n = 6 fields per group.

4. Discussion

SC

The current data showed that FCPR16 reversed CUMS-induced depressant-like behaviors in mice, accompanied by an upregulation of cAMP/CREB/BDNF and cAMP/EPAC signaling pathways as well as

NU

the expression of synapsin 1 and PSD95, an improvement in neurogenesis, and downregulation of neuroinflammation. To our knowledge, this is the first report that the PDE4 inhibitor FCPR16 exerted an

MA

antidepressant-like effect in CUMS-exposed mice. The idea is supported by the following results: (1) FCPR16 treatment ameliorated depressant-like behaviors in mice models of both acute stress and CUMS

D

without influencing the locomotor activity; (2) treatment with FCPR16 enhanced the activation of

TE

cAMP/CREB/BDNF and cAMP/EPAC signaling pathways; (3) FCPR16 improved CUMS-induced decreases in the expression of synapsin 1 and PSD95; (4) FCPR16 promoted neurogenesis in the

EP

hippocampus of CUMS mice; (5) pro-inflammatory cytokines were decreased while anti-inflammatory cytokines were increased with FCPR16 treatment; and (6) FCPR16 promoted a shift of microglial

AC C

phenotypes from M1 to M2.

In the behavior tests, OFT was used to evaluate locomotor activity in rodents (Wang et al., 2017). The durations were different among different labs, such as 60 min (Zanos et al., 2016), 20 min (Plattner et al., 2015) or 10 min (Zhang et al., 2016). However, we also noticed that 5-6 min of OFT was also commonly used in most labs (Cunha et al., 2017; Han et al., 2018; Zhao et al., 2018). In the present study, 6 min of OFT was used to rule out false positive results in the TST and FST due to hyperlocomotion (Siteneski et al., 2018), which was similar to our method for the OFT. In the TST, we occasionally found that a few

ACCEPTED MANUSCRIPT

mice grabbed their tails during the testing process. Under this condition, mice usually showed no intent to struggle, and this behavior is different than the immobility measured in the TST. Hence, mice showing

PT

tail-grabbing behaviors were not included in the final analysis (Can et al., 2012). Substantial evidence has revealed that PDE4 inhibitors exerted antidepressant-like effects via the

RI

cAMP/CREB/BDNF signaling pathway (Li et al., 2009; Bollen and Prickaerts, 2012). Using multiple biochemical and pharmacologic techniques, we observed increased cAMP concentrations upon FCPR16

SC

treatment in both the cerebral cortex and hippocampus and subsequent activation of CREB and EPAC-2. Based on these findings, we speculated that in addition to the cAMP/CREB/BDNF pathway, the

NU

cAMP/EPAC signaling pathway also contributed to the antidepressant-like effect of FCPR16. Due to the selective distribution of EPAC proteins, the effects of EPAC-2 on the CNS systems are

MA

foreseeable (Zhou et al., 2016). Previous studies (Srivastava et al., 2012; Yang et al., 2012) suggested that dendritic spine motility and cortical neuron density were reduced and the learning, memory and social

D

interaction functions were influenced in EPAC-2-deficient mice. In the current study, CUMS reduced the

TE

concentration of cAMP and decreased the expression level of EPAC-2 in the cerebral cortex and hippocampus. FCPR16 reversed these changes and improved the phosphorylation of ERK1/2. Summarily,

EP

these findings suggested that FCPR16 produced an antidepressant-like effect by upregulating the cAMP/CREB/BDNF and cAMP/EPAC signaling pathways.

AC C

In keeping with previous studies, synaptic damage and malfunction were accompanied by depression, which exhibited a decline in expression levels of synapsin 1 and PSD95 (Qiao et al., 2017). Our results showed that the expression levels of synapsin 1 and PSD95 were decreased in the cerebral cortex and hippocampus of CUMS-exposed mice and were significantly increased after FCPR16 treatment. With FCPR16 treatment, the number of DCX+ cells was increased in the hippocampus, which indicated that FCPR16 potentially improved neurogenesis in the hippocampus of mice exposed to CUMS. Depression and a variety of other psychiatric diseases are typically accompanied by neuroinflammation

ACCEPTED MANUSCRIPT

(Maes et al., 2009; Blandini, 2013; Heneka et al., 2014). Inhibiting inflammation and decreasing the levels of pro-inflammatory cytokines could relieve depressive symptoms (Kohler and Benros et al., 2014).

PT

In this study, we found that FCPR16 had an anti-neuroinflammation effect under chronic stress conditions, as reflected by decreased pro-inflammatory cytokines and increased transcription levels of M2 microglial

RI

markers.

Escitalopram produces antidepressant effects by increasing extracellular 5-hydroxytryptamine (5-HT).

SC

Elevated extracellular 5-HT activates G protein-coupled receptors and activates adenylyl cyclase. Escitalopram promotes the cAMP signaling pathway by accelerating generation instead of inhibiting the

NU

metabolism of cAMP. A previous study showed that simulation of the 5-HT7 receptor could suppress the release of pro-inflammatory cytokines (de Las et al., 2013). However, the results showed that

MA

escitalopram did not have a clear advantage in shifting M1/M2 microglial phenotypes. The expressions of pro-inflammatory cytokines and the mRNA levels of microglial M1 markers were decreased after

D

escitalopram treatment, but the levels of microglial M2 markers did not change significantly. We

TE

suspected that these results may be related to several issues. First, the regulation of microglial phenotypes is a dynamic process. Different drugs have different latent periods for regulating microglial polarizations

EP

(Zhang et al., 2017). The peak effect of escitalopram on microglial polarizations may not occur at the third week. Second, the role of escitalopram in cAMP is mainly to promote generation. Whether

AC C

increasing the cAMP level activates PDE4 is unknown. Most importantly, extracellular 5 -HT activates both 5-HT1A and 5-HT2A receptors, which will produce distinct or even opposing effects (Weisstaub et al., 2006; Isom et al., 2013; Hiroaki-Sato et al., 2014; Amidfar et al., 2017). The 5-HT1A receptor is expressed on activated T cells and regulates T cells proliferation (Aune et al., 1993). In contrast, activating the 5-HT2A receptor aggravates the inflammatory response both in peripheral tissues and CNS systems (Ito et al., 2000; Savignac et al., 2016). The latency periods of activating 5-HT1A and 5-HT2A receptors by escitalopram may be discrepant. This may be one of the reasons that less than 50% patients respond to

ACCEPTED MANUSCRIPT

SSRIs. However, there are also several insufficiencies in our experiment. Although FCPR16 has an

PT

antidepressant-like effect in mice, whether it influences the neurotransmitter involved in the pathology of depression is uncertain. It is deficient to evaluate synapsis function only by measuring the expression of

RI

synapse-associated proteins without measuring synaptic function by electrophysiology. Our studies have demonstrated that FCPR16 could protect neurons against apoptosis in MCAO rats and reverse

SC

depressant-like behaviors in CUMS mice. We investigated the changes in expression of related proteins in the cerebral cortex and hippocampus, brain regions closely associated with depression. However, we did

NU

not measure the FCPR16 distribution in specific brain regions. The above deficiencies remain to be

MA

further studied.

5. Conclusion

D

In summary, the current study demonstrated that the novel PDE4 inhibitor FCPR16 possessed an

TE

antidepressant-like effect in CUMS-exposed mice. The observed behavioral effects were apparently associated with the upregulation of cAMP/CREB/BDNF and cAMP/EPAC signaling pathways, as well as

EP

the expression of synapsin 1 and PSD95, improvement of neurogenesis, suppression of pro-inflammatory cytokine release and a shift in microglial M1/M2 polarization. Taken together, because of the

AC C

antidepressant-like effect of FCPR16 without an emesis side effect, FCPR16 could be a leading compound for new possible antidepressant research and development.

Disclosures

The authors declare that there is no conflict of interest.

ACCEPTED MANUSCRIPT

Ethical Statement All animal experiments performed in studies were in accordance the NIH Guide for the Care and Use of

PT

Laboratory Animals (NIH, Eighth Edition, 2011) and approved by the Laboratory Animal Ethics Committee of Southern Medical University (number 2016-0041). This article did not contain any studies

RI

with human performed by any of the authors.

SC

Acknowledgements

This work was supported by the National Nature Science Foundation of China (grant No: 81503043,

NU

81373384) and the Funding from Guangzhou Science and Technology Department (grant No.

MA

2015B020211007).

References

D

Amidfar M., Kim Y.K., Colic L., et al. 2017. Increased levels of 5HT2A receptor mRNA expression in peripheral blood mononuclear cells of patients with major depression: correlations with severity and

TE

duration of illness. Nord J Psychiatry. 71(4): 282-288. Aune T.M., McGrath K.M., Sarr T., et al. 1993. Expression of 5HT1a receptors on activated human T

151(3): 1175-1183.

EP

cells. Regulation of cyclic AMP levels and T cell proliferation by 5-hydroxytryptamine. J Immunol.

AC C

Blandini F. 2013. Neural and immune mechanisms in the pathogenesis of Parkinson's disease. J Neuroimmune Pharmacol. 8(1): 189-201. Bollen E., Prickaerts J. 2012. Phosphodiesterases in neurodegenerative disorders. IUBMB Life. 64(12): 965-970.

Can, A., Dao, D.T., Terrillion, C.E., et al. 2012. The Tail Suspension Test. J. Vis. Exp. (59), e3769, doi:10.3791/3769. Castagne V., Moser P., Roux S., et al. 2011. Rodent models of depression: forced swim and tail suspension behavioral despair tests in rats and mice. Curr Protoc Neurosci. Chapter 8. Unit 8.10A. Chen J., Yu H., Zhong J., et al. 2017. The phosphodiesterase-4 inhibitor, FCPR16, attenuates

ACCEPTED MANUSCRIPT

ischemia-reperfusion injury in rats subjected to middle cerebral artery occlusion and reperfusion. Brain Res Bull. 137: 98-106. Conti M., Beavo J. 2007. Biochemistry and physiology of cyclic nucleotide phosphodiesterases: essential components in cyclic nucleotide signaling. Annu Rev Biochem. 76: 481-511.

PT

Cui J., Jiang L., Xiang H. 2012. Ginsenoside Rb3 exerts antidepressant-like effects in several animal models. J Psychopharmacol. 26(5): 697-713.

RI

Cunha M.P., Pazini F.L., Lieberknecht V., et al. 2017. MPP(+)-Lesioned Mice: an Experimental Model of Motor, Emotional, Memory/Learning, and Striatal Neurochemical Dysfunctions. Mol Neurobiol. 54(8):

SC

6356-6377.

Dalla C., Antoniou K., Papadopoulou-Daifoti Z., et al. 2004. Oestrogen-deficient female aromatase

NU

knockout (ArKO) mice exhibit depressive-like symptomatology. Eur J Neurosci. 20(1): 217-228. de Las C.M., Dominguez-Soto A., Sierra-Filardi E., et al. 2013. Serotonin skews human macrophage polarization through HTR2B and HTR7. J Immunol. 190(5): 2301-2310.

MA

Fink G., Sumner B.E., Rosie R., et al. 1996. Estrogen control of central neurotransmission: effect on mood, mental state, and memory. Cell Mol Neurobiol. 16(3): 325-344. Galea L.A., Wide J.K., Barr A.M. 2001. Estradiol alleviates depressive-like symptoms in a novel animal

D

model of post-partum depression. Behav Brain Res. 122(1): 1-9.

TE

Garcia A.M., Martinez A., Gil C. 2016. Enhancing cAMP Levels as Strategy for the Treatment of Neuropsychiatric Disorders. Curr Top Med Chem. 16(29): 3527-3535. Geddes J.R., Miklowitz D.J. 2013. Treatment of bipolar disorder. Lancet. 381(9878): 1672 -1682.

EP

Han X., Wu H., Yin P., et al. 2018. Electroacupuncture restores hippocampal synaptic plasticity via modulation of 5-HT receptors in a rat model of depression. Brain Res Bull. 139: 256-262.

AC C

Heneka M.T., Kummer M.P., Latz E. 2014. Innate immune activation in neurodegenerative disease. Nat Rev Immunol. 14(7): 463-477. Hiroaki-Sato V.A., Sales A.J., Biojone C., et al. 2014. Hippocampal nNOS inhibition induces an antidepressant-like effect: involvement of 5HT1A receptors. Behav Pharmacol. 25(3): 187-196. Howren M.B., Lamkin D.M., Suls J. 2009. Associations of depression with C-reactive protein, IL-1, and IL-6: a meta-analysis. Psychosom Med. 71(2): 171-186. Isaac R., Boura-Halfon S., Gurevitch D., et al. 2013. Selective serotonin reuptake inhibitors (SSRIs) inhibit insulin secretion and action in pancreatic beta cells. J Biol Chem. 288(8): 5682-5693. Isom A.M., Gudelsky G.A., Benoit S.C., et al. 2013. Antipsychotic medications, glutamate, and cell death:

ACCEPTED MANUSCRIPT

a hidden, but common medication side effect? Med Hypotheses. 80(3): 252-258. Ito T., Ikeda U., Shimpo M., et al. 2000. Serotonin increases interleukin-6 synthesis in human vascular smooth muscle cells. Circulation. 102(20): 2522-2527. Kawasaki H., Springett G.M., Mochizuki N., et al. 1998. A family of cAMP-binding proteins that directly

PT

activate Rap1. Science. 282(5397): 2275-2279.

Kohler O., Benros M.E., Nordentoft M., et al. 2014. Effect of anti-inflammatory treatment on depression,

RI

depressive symptoms, and adverse effects: a systematic review and meta-analysis of randomized clinical trials. JAMA Psychiatry. 71(12): 1381-1391.

SC

Krishnan V., Nestler E.J. 2008. The molecular neurobiology of depression. Nature. 455(7215): 894-902. Kubera M., Obuchowicz E., Goehler L., et al. 2011. In animal models, psychosocial stress-induced (neuro)

NU

inflammation, apoptosis and reduced neurogenesis are associated to the onset of depression. Prog Neuropsychopharmacol Biol Psychiatry. 35(3): 744-759.

Lakics V., Karran E.H., Boess F.G. 2010. Quantitative comparison of phosphodiesterase mRNA

MA

distribution in human brain and peripheral tissues. Neuropharmacology. 59(6): 367-374. Li D.D., Xie H., Du YF, et al. 2017. Antidepressant-like effect of zileuton is accompanied by hippocampal

neuroinflammation

reduction

and

CREB/BDNF

upregulation

in

D

lipopolysaccharide-challenged mice. J Affect Disord. 227: 672-680.

TE

Li Y.C., Wang F.M., Pan Y., et al. 2009. Antidepressant-like effects of curcumin on serotonergic receptor-coupled AC-cAMP

pathway in chronic

unpredictable

mild stress

of

rats. Prog

Neuropsychopharmacol Biol Psychiatry. 33(3): 435-449.

EP

Li Y.F., Huang Y., Amsdell S.L., et al. 2009. Antidepressant- and anxiolytic-like effects of the phosphodiesterase-4 inhibitor rolipram on behavior depend on cyclic AMP response element binding

AC C

protein-mediated neurogenesis in the hippocampus. Neuropsychopharmacology. 34(11): 2404-2419. Liebenberg N., Muller H.K., Fischer C.W., et al. 2011. An inhibitor of cAMP -dependent protein kinase induces behavioural and neurological antidepressant-like effects in rats. Neurosci Lett. 498(2): 158-161. MacQueen G., Frodl T. 2011. The hippocampus in major depression: evidence for the convergence of the bench and bedside in psychiatric research? Mol Psychiatry. 16(3): 252-264. Maes M., Yirmyia R., Noraberg J., et al. 2009. The inflammatory & neurodegenerative (I&ND) hypothesis of depression: leads for future research and new drug developments in depression. Metab Brain Dis. 24(1): 27-53. Okada M., Hayashi N., Kometani M., et al. 1997. Influences of ovariectomy and continuous replacement

ACCEPTED MANUSCRIPT

of 17beta-estradiol on the tail skin temperature and behavior in the forced swimming test in rats. Jpn J Pharmacol. 73(1): 93-96. Plattner F., Hayashi K., Hernandez A., et al. 2015. The role of ventral striatal cAMP signaling in stress-induced behaviors. Nat Neurosci. 18(8): 1094-1100.

PT

Price J.L., Drevets W.C. 2010. Neurocircuitry of mood disorders. Neuropsychopharmacology. 35(1): 192-216.

RI

Qiao H., An S.C., Xu C., et al. 2017. Role of proBDNF and BDNF in dendritic spine plasticity and depressive-like behaviors induced by an animal model of depression. Brain Res. 1663: 29-37.

SC

Read J., Cartwright C., Gibson K. 2014. Adverse emotional and interpersonal effects reported by 1829 New Zealanders while taking antidepressants. Psychiatry Res. 216(1): 67-73.

NU

Savignac H.M., Couch Y., Stratford M., et al. 2016. Prebiotic administration normalizes lipopolysaccharide (LPS)-induced anxiety and cortical 5-HT2A receptor and IL1-beta levels in male mice. Brain Behav Immun. 52: 120-131.

MA

Schmidt M., Dekker F.J., Maarsingh H. 2013. Exchange protein directly activated by cAMP (epac): a multidomain cAMP mediator in the regulation of diverse biological functions. Pharmacol Rev. 65(2): 670-709.

D

Scott A.I., Perini A.F., Shering P.A., et al. 1991. In-patient major depression: is rolipram as effective as

TE

amitriptyline? Eur J Clin Pharmacol. 40(2): 127-129. Siteneski A., Cunha M.P., Lieberknecht V., et al. 2018. Central irisin administration affords antidepressant-like effect and modulates neuroplasticity-related genes in the hippocampus and prefrontal

EP

cortex of mice. Prog Neuropsychopharmacol Biol Psychiatry. 84(Pt A): 294-303. Srivastava D.P., Jones K.A., Woolfrey K.M., et al. 2012. Social, communication, and cortical structural

AC C

impairments in Epac2-deficient mice. J Neurosci. 32(34): 11864-11878. Thakare V.N., Patil R.R., Oswal R.J., et al. 2017. Therapeutic potential of silymarin in chronic unpredictable

mild stress

induced

depressive-like

behavior

in

mice. J

Psychopharmacol.

269881117742666.

Uher R., Mors O., Rietschel M., et al. 2011. Early and delayed onset of response to antidepressants in individual trajectories of change during treatment of major depression: a secondary analysis of data from the Genome-Based Therapeutic Drugs for Depression (GENDEP) study. J Clin Psychiatry. 72(11): 1478-1484. Wang C., Yang X.M., Zhuo Y.Y., et al. 2012. The phosphodiesterase-4 inhibitor rolipram reverses

ACCEPTED MANUSCRIPT

Abeta-induced cognitive impairment and neuroinflammatory and apoptotic responses in rats. Int J Neuropsychopharmacol. 15(6): 749-766. Wang P., Liu Z., Chen H., et al. 2017. Exchange proteins directly activated by cAMP (EPACs): Emerging therapeutic targets. Bioorg Med Chem Lett. 27(8): 1633-1639.

PT

Wang Q., Timberlake M.N., Prall K., et al. 2017. The recent progress in animal models of depression. Prog Neuropsychopharmacol Biol Psychiatry. 77: 99-109.

World Health Organization (2017) Depression Fact sheet.

SC

anxiety-like behaviors in mice. Science. 313(5786): 536-540.

RI

Weisstaub N.V., Zhou M., Lira A., et al. 2006. Cortical 5-HT2A receptor signaling modulates

http://www.who.int/mediacentre/factsheets/fs369/en/ (accessed February 2017).

NU

Willner P. 1997. Validity, reliability and utility of the chronic mild stress model of depression: a 10 -year review and evaluation. Psychopharmacology (Berl). 134(4): 319-329. Willner P., Towell A., Sampson D., et al. 1987. Reduction of sucrose preference by chronic unpredictable

MA

mild stress, and its restoration by a tricyclic antidepressant. Psychopharmacology (Berl). 93(3): 358-364. Yang Y., Shu X., Liu D., et al. 2012. EPAC null mutation impairs learning and social interactions via aberrant regulation of miR-124 and Zif268 translation. Neuron. 73(4): 774-788.

D

Zanos P., Moaddel R., Morris P.J., et al. 2016. NMDAR inhibition-independent antidepressant actions of

TE

ketamine metabolites. Nature. 533(7604): 481-486. Zhang J.Q., Wu X.H., Feng Y., et al. 2016. Salvianolic acid B ameliorates depressive-like behaviors in

37(9): 1141-1153.

EP

chronic mild stress-treated mice: involvement of the neuroinflammatory pathway. Acta Pharmacol Sin.

Zhang J., Xie X., Tang M., et al. 2017. Salvianolic acid B promotes microglial M2 -polarization and

AC C

rescues neurogenesis in stress-exposed mice. Brain Behav Immun. 66: 111-124. Zhao Q., Peng C., Wu X., et al. 2014. Maternal sleep deprivation inhibits hippocampal neurogenesis associated with inflammatory response in young offspring rats. Neurobiol Dis. 68: 57 -65. Zhao Y., Li H., Fang F., et al. 2018. Geniposide improves repeated restraint stress-induced depression-like behavior in mice by ameliorating neuronal apoptosis via regulating GLP-1R/AKT signaling pathway. Neurosci Lett. 676: 19-26. Zhou L., Ma S.L., Yeung P.K., et al. 2016. Anxiety and depression with neurogenesis defects in exchange protein directly activated by cAMP 2-deficient mice are ameliorated by a selective serotonin reuptake inhibitor, Prozac. Transl Psychiatry. 6(9): e881.

ACCEPTED MANUSCRIPT

Zhou Z.Z., Ge B.C., Zhong Q.P., et al. 2016. Development of highly potent phosphodiesterase 4 inhibitors with anti-neuroinflammation potential: Design, synthesis, and structure-activity relationship study of catecholamides bearing aromatic rings. Eur J Med Chem. 124: 372-379. Zou Z.Q., Chen J.J., Feng H.F., et al. 2017. Novel Phosphodiesterase 4 Inhibitor FCPR03 Alleviates

PT

Lipopolysaccharide-Induced Neuroinflammation by Regulation of the cAMP/PKA/CREB Signaling

AC C

EP

TE

D

MA

NU

SC

RI

Pathway and NF-kappaB Inhibition. J Pharmacol Exp Ther. 362(1): 67-77.

ACCEPTED MANUSCRIPT

Ethical Statement The authors declare there is no conflict of interest. All animal experiments performed in studies were in

PT

accordance the NIH Guide for the Care and Use of Laboratory Animals (NIH, Eighth Edition, 2011) and approved by the Laboratory Animal Ethics Committee of Southern Medical University (number

AC C

EP

TE

D

MA

NU

SC

RI

2016-0041). This article did not contain any studies with human performed by any of the authors.

ACCEPTED MANUSCRIPT

Highlights FCPR16 ameliorates depressant-like behaviors in CUMS-exposed mice.



Treatment of FCPR16 up-regulates cAMP-mediated signaling pathways in mice.



FCPR16 increases the expression of synapsin1 and PSD95 in mice.



FCPR16 treatment suppresses the neuroinflammation in mice.

AC C

EP

TE

D

MA

NU

SC

RI

PT



Figure 1

Figure 2

Figure 3

Figure 4

Figure 5

Figure 6

Figure 7

Figure 8

Figure 9