Skip to main content

Ahi1 regulates serotonin production by the GR/ERβ/TPH2 pathway involving sexual differences in depressive behaviors

Abstract

Background

Depression is one of the most common psychiatric diseases. The monoamine transmitter theory suggests that neurotransmitters are involved in the mechanism of depression; however, the regulation on serotonin production is still unclear. We previously showed that Ahi1 knockout (KO) mice exhibited depression-like behavior accompanied by a significant decrease in brain serotonin.

Methods

In the present study, western blot, gene knockdown, immunofluorescence, dual-luciferase reporter assay, and rescue assay were used to detect changes in the Ahi1/GR/ERβ/TPH2 pathway in the brains of male stressed mice and male Ahi1 KO mice to explain the pathogenesis of depression-like behaviors. In addition, E2 levels in the blood and brain of male and female mice were measured to investigate the effect on the ERβ/TPH2 pathway and to reveal the mechanisms for the phenomenon of gender differences in depression-like behaviors.

Results

We found that the serotonin-producing pathway-the ERβ/TPH2 pathway was inhibited in male stressed mice and male Ahi1 KO mice. We further demonstrated that glucocorticoid receptor (GR) as a transcription factor bound to the promoter of ERβ that contains glucocorticoid response elements and inhibited the transcription of ERβ. Our recent study had indicated that Ahi1 regulates the nuclear translocation of GR upon stress, thus proposing the Ahi1/GR/ERβ/TPH2 pathway for serotonin production. Interestingly, female Ahi1 KO mice did not exhibit depressive behaviors, indicating sexual differences in depressive behaviors compared with male mice. Furthermore, we found that serum 17β-estradiol (E2) level was not changed in male and female mice; however, brain E2 level significantly decreased in male but not female Ahi1 KO mice. Further, ERβ agonist LY-500307 increased TPH2 expression and 5-HT production. Therefore, both Ahi1 and E2 regulate the ERβ/TPH2 pathway and involve sexual differences in brain serotonin production and depressive behaviors.

Conclusions

In conclusion, although it is unclear how Ahi1 controls E2 secretion in the brain, our findings demonstrate that Ahi1 regulates serotonin production by the GR/ERβ/TPH2 pathway in the brain and possibly involves the regulation on sex differences in depressive behaviors.

Video Abstract

Background

Brain serotonin (5-hydroxytryptamine, 5-HT) plays an essential role in different brain functions, including modulating brain development, stress reactivity, cognition, sleep, and aggressive behaviors; the changes in brain serotonergic function may be related to different symptoms in patients with MDD that are associated with the physiological processes of serotonin [1,2,3,4,5,6,7]. Therefore, maintenance of brain serotonin homeostasis is critical for normal behaviors and mental health. Brain serotonin is mainly produced in the serotonergic raphe nuclei because tryptophan hydroxylase 2 (TPH2), the rate-limiting enzyme of serotonin synthesis, is highly expressed in this region. Increasing evidence indicates that TPH2 is related to MDD in depression [8,9,10]. A previous study demonstrated that estrogen receptor-β (ERβ) binds an estrogen response element in the promoter of TPH2 and regulates the expression of TPH2 mRNA [11], which may connect serotonin, TPH2, and ERβ. Our previous study showed that Ahi1 deficiency causes a reduction in serotonin levels in multiple brain regions and depressive behaviors in mice [12]; however, whether Ahi1 regulates brain serotonin levels through the ERβ/TPH2/serotonin pathway is unclear.

Glucocorticoid receptor (GR), a ligand-dependent regulatory transcription factor, can repress or induce the transcription of thousands of genes by directly binding to DNA response elements and/or other transcription factors [13]. In addition, our recent results indicate that AHI1 interacts directly with the C-terminus of GR to inhibit GR nuclear translocation [14]. Therefore, we speculate that GR may be involved in the regulation of serotonin by Ahi1.

Increasing evidence indicates that 17β-estradiol (E2) is involved in regulating depression, and estrogen supplementation can alleviate the depressive symptoms of patients [15, 16]. In animal studies, E2 treatment was also shown to ameliorate depressive behavior [17, 18]. In addition, there is growing evidence that E2 exerts an antidepressant effect through ERβ but not ERα [19] and that ERβ agonists also have antidepressant-like effects [20, 21]. More interestingly, the antidepressant-like effects of E2 are absent in ERβ knockout (KO) mice [22]. It has been reported that estradiol profoundly influences mood by acting on serotonergic function, but the specific mechanism is not clear [23, 24]. These data suggest that E2 may exert an antidepressant effect by regulating serotonin through ERβ.

Sex differences in vulnerability to many mental illnesses, such as depression, anxiety, alcoholism, and drug abuse, have been identified [25,26,27,28]. An increasing amount of evidence has demonstrated the existence of sex differences in major depressive disorders (MDD); there are sex differences not only in the clinical symptoms of depression [29] but also in prevalence, comorbidities, and antidepressant efficacy [30,31,32,33]. For example, the risk of depression is twice as high in adult females as in adult males [34, 35]. Interestingly, prior to puberty, there are no sex differences in the prevalence of MDD [36]. Therefore, sex differences in MDD, as the most prevalent psychiatric disorder, are becoming essential in this field, and the underlying mechanisms are being investigated to aid the development of personalized treatments for patients with MDD. Among various factors, sex differences in serotonergic function are considered a significant contributing factor to sex differences in MDD [37, 38]. Moreover, deficiencies in serotonin synthesis, transport, and degradation are modulated by sex differences [39, 40]. In addition, sex differences in gonadal hormones and HPA axis activation modulate sex differences in depression [41,42,43,44]. The process is complicated because how these factors interact is still largely unknown.

In this study, we found that Ahi1, a protein associated with susceptibility to depression [14, 45], regulated serotonin production through the GR/ERβ/TPH2 pathway, indicating a critical role for Ahi1 in the regulation of depression. Sex differences in brain estrogen levels may at least partially account for the differences in depressive behaviors between male and female Ahi1 KO mice, indicating that Ahi1 regulates serotonin production by the GR/ERβ/TPH2 pathway involving sexual differences in depression-like behaviors.

Materials and methods

Antibodies and reagents

ERβ (ab3576), ERα (ab32063), TPH2 (ab184505), SERT (ab102048), and Histone3 (ab1791) antibodies were purchased from Abcam Company (Abcam, MA, USA); Ahi1 (sc-515382) and GR (sc-56851) antibodies were obtained from Santa Cruz Biotechnology Inc. (Santa Cruz, CA, USA); a mouse estradiol ELISA kit was purchased from Cusabio Biological Engineering Co., Ltd. (Baltimore, MD, USA); 17β-estradiol was purchased from Sigma-Aldrich Company (St. Louis, MO, USA); a Transcriptor First Strand cDNA Synthesis Kit and 2XSYBR Green PCR Master Mix were obtained from Roche (Germany); and Lipofectamine 2000 was purchased from Invitrogen Corporation (San Diego, CA, USA). All secondary antibodies were obtained from Jackson ImmunoResearch Laboratories (West Grove, PA, USA).

Mice

Healthy female and male C57BL/6 J mice (23–25 g) were purchased from SLAC Company (Shanghai, China). Ahi1KO mice were described previously [12, 46]. Mice were randomly housed in polypropylene cages at 22 ± 2 °C under a 12 h:12 h light–dark cycle with free access to food and water. All behavioral tests were conducted in the light phase between 9:00 and 12:00 am. The mice were brought to the testing room approximately 30 min before the test, temporarily placed in a cage after each test, and then returned to their original cage. There was an interval of at least 24 h between successive behavioral tests. All experiments were performed in accordance with the guidelines of the University Committee on Animal Care of Soochow University.

Spatial restraint stress

A spatial restraint stress model was generated as described previously [47]. Adult C57BL/6 J mice were randomly divided into the control group and the stress group. The mice in the stress group were individually placed into a well-ventilated 50 ml centrifuge tube for 2 h (from 9:00 to 11:00) daily to restrict the movements of the mice. During spatial restraint, the mice were deprived of food and water. The mice were freely housed in their original cages in the control group but were not given access to food and water. After 2 h of spatial restraint, the mice were placed back in their original cages and given food and water. The behavioral tests were performed every 7 d.

Forced swimming test (FST)

The FST was carried out as described previously [48]. Each mouse was placed in a 2.0 L glass beaker (diameter 10 cm, height 25 cm) filled with 14 cm of water (25 °C) and forced to swim for 6 min. Immobility time (floating while making the minimum movements necessary to keep the head above water) in 6 min was recorded. The mice were wiped with a paper towel to keep them warm and gently dried in their home cages for 30 min. After each trial, the water in the cylinder was replaced.

Tail suspension test (TST)

The TST was performed as described previously with slight modifications [47]. In this experiment, the tail tip of each mouse was fixed to a wooden rod with adhesive tape, and the mouse was suspended 35 cm above a desk. A stopwatch was used to record the immobility time in a 6 min period. The mouse was placed back in its original cage at the end of each trial.

Measurement of serotonin (5-HT) levels

The serotonin content in the hippocampus was measured by high-performance liquid chromatography (HPLC) as described previously [12]. Hippocampal tissues were homogenized with an ultrasonic homogenizer in 200 μl 0.4 M perchloric acid. The homogenates were centrifuged at 10,000×g for 15 min. After 0.4 M perchloric acid was added to the supernatants to a volume of 1 ml, the samples were then injected into the HPLC system. The serotonin levels in the samples were measured according to a serotonin standard [12, 47].

Western blot analysis

Mice were killed, and different brain tissues, including the hippocampus and the brainstem, were collected. The samples were homogenized in lysis buffer, and the homogenates were centrifuged at 4 °C. The supernatants were collected, and the protein concentrations were determined. The protein samples (20–40 μg) were separated by 10% or 12% sodium dodecyl sulfate–polyacrylamide gel electrophoresis and subsequently transferred to nitrocellulose membranes. The membranes were blocked with 5% milk in phosphate-buffered saline/0.1% Tween 20 (PBST) for 1 h; then, the membranes were incubated with primary antibodies with shaking at 4 °C overnight. The membranes were incubated with horseradish peroxidase-conjugated secondary antibody for 1 h at room temperature on the second day. After washing, the protein signals were developed with an ECL chemiluminescence system, and densitometric analysis of the bands was carried out with Alpha Ease Image Analysis Software (version 3.1.2, Alpha Innotech).

Immunofluorescence staining

Immunofluorescence staining was performed as previously described [49]. Mice were perfused with ice-cold saline and 4% paraformaldehyde via the heart. After the brains were immersed in 15% and 30% sucrose solution (w/v) for 24 h, the brain tissues were cut into Sects. (12 μm) with a freezing cryostat. The brain slices were attached to glass slides and washed with phosphate-buffered saline (PBS) for 30 min. The slices were incubated at room temperature with buffer containing 0.3% Triton-X-100/5% BSA/PBS for 1 h. The sections were incubated with primary antibodies overnight at 4 °C. On the second day, the sections were incubated with secondary antibodies and the nuclear dye DAPI for 1 h at 4 °C. Images were captured with a fluorescence microscope (Axio ScopeA1, Carl Zeiss, Oberkochen, Germany).

Quantitative PCR (Q-PCR) analysis

Total RNA was isolated from brain tissues or cells by using an RNeasy Plus Mini kit according to the manufacturer’s instructions. cDNA was synthesized by reverse transcription using the Transcriptor First Strand cDNA Synthesis Kit as described previously [50, 51]. Real-time PCR was performed using a 7500 real-time PCR machine (Applied Biosystems, USA) in a volume of 20 μl, which consisted of 50 ng cDNA, 10 μl of 2XSYBR Green PCR Master Mix, and 1 μl primers (10 μM). GAPDH was used as an internal loading control. The PCR primers are listed in Additional file 1: Table S1.

Estradiol measurement by ELISA

After the mice were killed, different brain tissues and plasma samples were collected. According to the instructions, the estradiol content in brain tissues and plasma was determined using a commercial estradiol ELISA kit (Cusabio Biological Engineering Co., Ltd., Baltimore, MD, USA).

Plasmid construction

The ERβ promoter region was isolated by PCR amplification of human cDNA, and then the cDNAs were inserted into the pGL3-basic vector. A plasmid containing a 2-kb region of the ERβ promoter followed by a luciferase reporter gene was transfected into 293 T cells that were treated with Dex and/or GR-siRNA for 48 h. Luciferase activity was measured using a Dual-Luciferase Assay Kit (Promega, USA) according to the manufacturer's protocol. The assay results were presented as relative luciferase activity.

Cell culture and plasmid transfection

PC12 cells or 293 T cells were maintained in DMEM (HyClone, Logan, UT, USA) containing 10% fetal bovine serum and 100 μg/ml streptomycin sulfate. When the density of the PC12 cells or 293 T cells reached approximately 50–60%, plasmid transfection was carried out. Plasmid or siRNA was mixed with 50 μl serum-free medium. Then, 2 μl Lipofectamine 2000 (Invitrogen Corporation, City, USA) was mixed with 50 μl serum-free medium. Plasmid DNA or siRNA was further mixed with Lipofectamine 2000 and incubated for 15 min at room temperature. The mixture (100 μl) was added to each well of a 6-well plate containing 900 μl serum-free medium. After 6 h, the transfection medium was removed and replaced with regular culture medium. After 48 h, cells were collected for further Western blot analysis.

Nuclear and cytoplasmic extraction

Fresh cells or brain tissues were collected for further experiments, and the nuclear and cytoplasmic fractions were separated with a nuclear and cytoplasmic protein extraction kit (Beyotime Biotechnology, P0028, Shanghai, China). The nuclear and cytoplasmic fractions were used for further Western blot analysis.

Administration of drugs to animals

The ERβ agonist LY500307 (Selleckchem, Houston, TX, USA) was dissolved in DMSO and freshly prepared daily before treatment. Ahi1 KO mice were randomly divided into five groups (N = 9–10 mice in each group): the vehicle-treated control group, the control treated with the ERβ agonist LY500307 (0.2 mg/kg), the vehicle-treated Ahi1 KO group, the Ahi1 KO group treated with low-dose LY500307 (0.05 mg/kg), and the Ahi1 KO group treated with high-dose LY500307 (0.2 mg/kg). The mice were treated intraperitoneally for three weeks, and behavioral tests were performed after treatment with LY500307.

Statistical analysis

The data were presented as the mean ± SEM. Differences between two groups were analyzed by Student’s t test. Differences among groups were analyzed by one-way ANOVA followed by the LSD post hoc test or two-way ANOVA followed by Bonferroni’s multiple comparisons post hoc test where appropriate. A value of p < 0.05 was considered significant.

Results

The ERβ/TPH2/5-HT pathway was inhibited in male Ahi1 KO mice and stressed mice

Our previous studies demonstrated that Ahi1 knockout causes depressive phenotypes in male mice [12, 46, 52]. For males, the immobility time of Ahi1 KO mice was significantly increased compared with that of control mice (Fig. 1A, B). To explore the underlying mechanisms of depression-like behaviors, we measured 5-HT levels by HPLC in hippocampal tissues from male Ahi1 KO mice because previous studies have shown that serotonin is involved in the regulation of depression [12]. The 5-HT level was significantly decreased in the hippocampal tissues of male Ahi1 KO mice (Fig. 1C), suggesting that serotonin levels may cause depressive behaviors in male Ahi1 KO mice. We further explored the regulatory pathway of 5-HT production. Although MAO activity partially affects total 5-HT levels in tissues [12], determinant factors, including 5-HT transporter SERT and the limiting enzyme for 5-HT production TPH2 are related to the recycling and production of 5-HT, respectively [11]. Our findings showed that Ahi1 KO did not alter SERT expression (Fig. 1D); however, TPH2 mRNA levels in the hippocampus (Fig. 1E) and the brainstem (Additional file 2: Fig. S1B) significantly decreased in male Ahi1 KO mice. Similarly, TPH2 protein expression in the hippocampus (Fig. 1F) and the brainstem (Additional file 2: Fig. S1A) in male Ahi1 KO mice significantly decreased compared with that in control mice. Consistently, ERβ mRNA levels in the hippocampus (Fig. 1E) and brainstem (Additional file 2: Fig. S1C) were significantly reduced in male Ahi1 KO mice. ERβ protein levels in the hippocampus (Fig. 1G) and the brainstem (Additional file 2: Fig. S1D) were markedly decreased in male Ahi1 KO mice compared with male control mice. Immunofluorescence staining also confirmed the reduction in ERβ protein expression in the male Ahi1 KO mouse brain (Fig. 1H), but the ERβ positive cells had no difference between the two groups (Additional file 2: Fig. S1F). The protein expression of ERα, an important isoform of ER, also did not change in the hippocampi of Ahi1 KO mice (Additional file 2: Fig. S1E). Therefore, the ERβ/TPH2 pathway was affected in male Ahi1 KO mice.

Fig. 1
figure 1

The ERβ/TPH2/5-HT pathway was down-regulated in male Ahi1 KO mice. The immobility time in TST (A) and FST (B) were performed in healthy male 2–3 month-old Ahi1 KO mice. N = 7–9. C The content of 5-HT was determined by HPLC in hippocampus samples from male Ahi1 KO mice. N = 6. D SERT protein expression was examined in hippocampus samples from male Ahi1 KO mice by Western blot analysis. N = 3. E TPH2 and ERβmRNA levels were quantified by quantitative PCR in the hippocampus tissues of male Ahi1 KO mice. N = 3–5. F TPH2 protein expression was examined in hippocampus tissues from male Ahi1 KO mice by Western blot analysis. N = 3. G ERβ protein expression was examined in hippocampus tissues from male Ahi1 KO mice by Western blot analysis. N = 3. H ERβ protein expression was detected in the brainstem in male Ahi1 KO mice by fluorescent staining. Scale bar = 20 μm. *p < 0.05; **p < 0.01; ****p < 0.0001 versus Control (Con)

To further confirm that the ERβ/TPH2/5-HT pathway is also changed in male stressed mice, spatial restraint stress was used to induce depressive behaviors in mice. The immobility time in TST and FST was significantly increased for male stressed mice after spatial restraint stress for 2 w compared with control male mice (Additional file 3: Fig. S2A, B). Similarly, TPH2 mRNA and protein levels (Additional file 4: Fig. S3A, B) were also decreased markedly in the hippocampus in male stressed mice compared with male control mice. ERβ mRNA and protein (Additional file 4: Fig. S3C, D) levels were significantly reduced in male stressed mice. SERT expression in male stressed mice also did not change (Additional file 4: Fig. S3E). Importantly, Ahi1 protein expression decreased significantly in male stressed mice (Additional file 3: Fig. S2C), suggesting that Ahi1 is critical for the ERβ/TPH2/5-HT pathway in regulating depressive behaviors.

Ahi1 positively regulated ERβ expression

To examine the regulatory effect of Ahi1 on the ERβ/TPH2/5-HT pathway, we knocked down Ahi1 in PC12 cells and examined ERβ expression. After Ahi1-siRNA transfection for 48 h, Ahi1 was significantly knocked down (Fig. 2A). ERβ protein (Fig. 2A) and mRNA levels (Fig. 2B) were both significantly decreased. Because ERβ is one of the classical nuclear receptors of estrogen and is mainly distributed in the nucleus [53, 54], we further determined the effect of Ahi1 on cellular ERβ distribution. Ahi1 knockdown reduced ERβ expression in the nucleus but not in the cytoplasm (Fig. 2C). Consistently, male Ahi1 KO mice showed a significant decrease in nuclear ERβ expression in the hippocampus; however, cytoplasmic ERβ expression was not significantly changed by Ahi1 KO (Fig. 2D). These findings suggested that Ahi1 knockdown or KO not only decreased ERβ expression but also reduced the nuclear translocation of ERβ, hindering its functions as a nuclear receptor.

Fig. 2
figure 2

Ahi1 positively regulated ERβ expression. A, Ahi1 and ERβ protein expression were examined in Ahi1-siRNA transfected PC12 cells 48 h by Western blot analysis. N = 5. B ERβ mRNA level in Ahi1-siRNA transfected PC12 cells 48 h was examined by quantitative PCR Quantitative analysis. N = 3. C ERβ protein expression was detected in the cytoplasm and nucleus of Ahi1-siRNA transfected PC12 cells 48 h by Western blot analysis. N = 3. D ERβ protein expression was detected in cytoplasm and nucleus in the hippocampus tissue from male Ahi1 KO mice by Western blot analysis. N = 4. *p < 0.05; **p < 0.01; ***p < 0.001 versus Control

GR inhibited the expression of ERβ by binding to the promoter of ERβ

Previous studies have demonstrated that Ahi1 is expressed predominantly in the cytoplasm, neuronal processes, and sigmoid bodies but is found at lower levels in the nucleus [55, 56]. Therefore, we speculated that Ahi1 might regulate ERβ transcription indirectly by interacting with some transcription factors. Our recent results indicated that Ahi1 interacts directly with glucocorticoid receptor (GR), an effector of the HPA system, and inhibits GR nuclear translocation [14]. However, it is still unclear whether Ahi1 regulates ERβ expression through the transcriptional effects of GR. Therefore, we examined the cellular distribution of GR after Ahi1-siRNA transfection. GR protein expression was significantly reduced in the cytoplasm and increased in the nucleus (Fig. 3A), suggesting that Ahi1 has an inhibitory effect on GR nuclear translocation. We further examined the regulatory effect of increased GR nuclear translocation on ERβ expression. PC12 cells were treated with dexamethasone (Dex, an agonist of GR that promotes GR nuclear translocation). After Dex treatment for 72 h, ERβ protein (Fig. 3B) and mRNA (Fig. 3C) levels decreased significantly. Interestingly, nuclear ERβ protein expression was significantly reduced (Additional file 5: Fig. S4A), but cytoplasmic ERβ protein expression did not change (Additional file 5: Fig. S4B). In contrast, after GR was knocked down by GR-siRNA in PC12 cells, ERβ mRNA (Fig. 3D) and protein (Fig. 3E) levels increased significantly. Therefore, the results showed that increased GR nuclear translocation inhibited the transcription of ERβ mRNA.

Fig. 3
figure 3

GR inhibited the expression of ERβ by binding to the promoter of ERβ. A GR protein expression was detected in the cytoplasm and nucleus of Ahi1-siRNA transfected PC12 cells by Western blot analysis. N = 3. B ERβ protein expression was detected in Dex treated PC12 cells by Western blot analysis. N = 3. C ERβ mRNA levels in Dex treated PC12 cells were examined by quantitative PCR Quantitative analysis. N = 3–4. D ERβ mRNA levels in GR-siRNA transfected PC12 cells were examined by quantitative PCR Quantitative analysis. N = 3. E GR and ERβ protein expression were examined in GR-siRNA transfected PC12 cells by Western blot analysis. N = 3. F PGL3-basic-ERβ-promoter Plasmid transfected 293 T cells were treated with Dex and/or GR-siRNA, and the luciferase activity was measured using a Dual-Luciferase Assay kit. N = 5. *p < 0.05; **p < 0.01; ****p < 0.0001 versus Control

To clearly demonstrate the direct regulatory effect of GR on ERβ mRNA transcription, we constructed a plasmid that contained a 2-kb region of the promoter of ERβ followed by a luciferase reporter gene. Plasmid-transfected 293 T cells were treated with Dex and/or GR-siRNA. In control-siRNA treated cells, luciferase expression was significantly reduced in cells treated with Dex compared with vehicle-treated cells (lane 1 and lane 2; Fig. 3F); however, compared with vehicle/control-siRNA, GR-siRNA increased luciferase expression (lane 1 and lane 3; Fig. 3F). Interestingly, Dex treatment did not reverse the GR knockdown-induced increase in luciferase expression (lane 3 and lane 4; Fig. 3F). A comparison of the two Dex-treated groups revealed that GR knockdown increased reporter gene expression (lane 2 and lane 4; Fig. 3F). These data demonstrated that GR could interact with the ERβ promoter and inhibits the transcription of ERβ mRNA.

An ERβ agonist reversed depression-like behaviors and increased TPH2 and serotonin production in male Ahi1 KO mice

Previous studies have demonstrated that ERβ agonists ameliorate depressive behaviors [20, 21, 57] and that the antidepressant-like effects of E2 are absent in ERβ knockout mice [22]. It is unclear whether ERβ agonists rescue depression-like behaviors in Ahi1 KO mice. Consistent with our previous study, male Ahi1 KO mice showed longer immobility times than control mice in the TST and FST (Fig. 4A, B). After treatment with the ERβ agonist LY-500307 (LY, 0.05 mg-0.2/kg/day) for 3 weeks, LY treatment at a higher dosage (0.2 mg/kg/day) had a significant antidepressant effect in the TST and FST (Fig. 4A, B). Compared with vehicle treatment, LY (0.2/kg/day) treatment reversed the decrease in 5-HT content (Fig. 4C) and TPH2 protein expression (Fig. 4D, E) in male Ahi1 KO mice.

Fig. 4
figure 4

ERβ agonist (LY-500307) reversed depression-like behaviors and increased TPH2 and serotonin production in male Ahi1 KO mice. A, B After 3 weeks treatment with exogenous ERβ agonist, behavioral tests were performed, the immobility time in TST and FST was recorded. N = 7–14. C After 3 weeks treatment with an exogenous ERβ agonist, the content of 5-HT in the hippocampus samples from male mice was determined by HPLC. N = 6. D, E TPH2 protein expression in the hippocampus samples from male Ahi1 KO mice was examined by Western blot analysis. N = 3. **p < 0.01; ***p < 0.001; ****p < 0.0001

The ERβ/TPH2/5-HT pathway was not changed in female Ahi1 KO mice and stressed mice

To show the effect of Ahi1 KO on depressive phenotypes in mice of different sexes, we also subjected female Ahi1 KO mice to behavioral tests, including the TST and FST. Interestingly, for females, the immobility time of Ahi1 KO mice was not different from that of control mice (Fig. 5A, B), indicating that there were sex differences in depressive phenotypes in Ahi1 KO mice. To explore the underlying mechanisms of the sex differences, we measured 5-HT levels by HPLC in hippocampal tissues from female Ahi1 KO mice because previous studies have shown that serotonin is involved in regulating sex differences in depression [58], hippocampal 5-HT levels in female Ahi1 KO mice were not different from those in control mice (Fig. 5C), consistent with its normal phenotype. This suggested that the difference in serotonin levels between the sexes may cause sex differences in depressive behaviors in Ahi1 KO mice. We further explored the changes in ERβ/TPH2/5-HT pathway in female Ahi1 KO mice. Our results showed no changes in TPH2 mRNA level and protein expression in the hippocampus (Fig. 5D–F) and the brainstem (Additional file 2: Fig. S1A, B) in female AHI1 KO mice. Consistently, ERβ mRNA level and protein expression in the hippocampus (Fig. 5G–I) and brainstem (Additional file 2: Fig. S1C, D) were also unaltered in female Ahi1 KO mice. Therefore, the ERβ/TPH2/5-HT pathway was affected in female Ahi1 KO mice.

Fig. 5
figure 5

The ERβ/TPH2/5-HT pathway was not changed in female Ahi1 KO mice. The immobility time in TST (A) and FST (B) were performed in healthy female 2–3 month-old Ahi1 KO mice. N = 7–9. C The content of 5-HT was determined by HPLC in hippocampus samples from female Ahi1 KO mice. N = 6. D TPH2 mRNA level was quantified by quantitative PCR in the hippocampus tissues from female Ahi1 KO mice. N = 3. E, F TPH2 protein expression was examined in hippocampus tissues from female Ahi1 KO mice by Western blot analysis. N = 3. G ERβ mRNA level was quantified by quantitative PCR in the hippocampus tissues from female Ahi1 KO mice. N = 3. H, I ERβ protein expression was examined in hippocampus tissues from female Ahi1 KO mice by Western blot analysis. N = 3

To further confirm whether the ERβ/TPH2/5-HT pathway is also unchanged in female stressed mice, spatial restraint stress was used to induce depressive behaviors in mice. The immobility time of female stressed mice was not different from that of female control mice (Additional file 3: Fig. S2A, B). In order to explain this sex difference, we measured Ahi1 protein and ERβ/TPH2/5-HT pathway. The Ahi1 protein level (Additional file 3: Fig. S2D) did not change in female stressed mice compared with male control mice. Similarly, TPH2 mRNA and protein levels (Additional file 4: Fig. S3A, B) were not altered in the hippocampus in female stressed mice compared with male control mice; ERβ mRNA and protein (Additional file 4: Fig. S3C, D) levels were also unchanged in female stressed mice. These results showed that the normal phenotype and 5-HT content of female AHI1 KO mice and stressed mice were not only affected by Ahi1/GR/ERβ/TPH2/5-HT pathway regulation but may also be related to other pathways.

Brain E2 expression was reduced in male Ahi1 KO mice but not in female Ahi1 KO mice.

Due to the therapeutic effect of the ERβ agonist LY in male Ahi1 KO mice, we explored whether the estrogen level in the brain or plasma affects sex differences in depressive behaviors. The contents of E2 in the brainstem and the hippocampus were not different between female Ahi1 KO mice and female control mice (Fig. 6A, B); however, the content of E2 in the brainstem and the hippocampus was significantly decreased in male Ahi1 KO mice compared with male control mice (Fig. 6A, B). The content of brain E2 was significantly higher in female mice than in male control mice (Fig. 6A, B). Interestingly, E2 levels in plasma were not different between female and male Ahi1 KO mice and their corresponding controls (Fig. 6C), and E2 levels in plasma were only slightly higher in female Ahi1 KO mice than in male Ahi1 KO mice (Fig. 6C). Therefore, our findings suggested that endogenous E2 in the brain may also be involved in the regulation of depressive behaviors through the ERβ/TPH2/5-HT pathway. In male Ahi1 KO mice, the reduction in 5-HT levels can be easily explained by the Ahi1/GR/ERβ/TPH2/5-HT pathway; however, this pathway does not explain the expected levels of ERβ, TPH2, and 5-HT in the brain tissues of female Ahi1 KO mice. Sex differences were explained by increased E2 levels in the brains of female Ahi1 KO mice because E2 may compete with GR for the ERβ promoter and alleviate the inhibition of the ERβ transcription by GR.

Fig. 6
figure 6

Brain E2 was reduced in male Ahi1 KO mice but not in female Ahi1 KO mice. A, B The content of E2 in the brainstem (A) and the hippocampus (B) from male and female Ahi1 KO mice was detected by ELISA kit. N = 6. C The content of E2 in plasma from female Ahi1 KO mice and male Ahi1 KO mice was also detected by an ELISA kit. N = 6. D The proposed diagram of the Ahi1 regulates serotonin production by the GR/ERβ/TPH2 pathway involved in sex differences in depressive behavior. In male Ahi1 KO mice, a reduction in Ahi1 expression decreases the cytoplasmic GR level, promotes ligand-dependent GR nuclear translocation and ERβ/TPH2/5-HT pathway was down-regulated; the lower E2 content in male mice could not compensate for the decrease in ERβ expression and depressive behavior in male Ahi1 KO mice; In female Ahi1 KO mice, high levels of E2 promoted ERβ protein expression and compensated for the Ahi1 deficiency-induced reduction in ERβ protein expression. Finally, these changes caused an increase in TPH2 and 5-HT levels, leading to normal behaviors in female Ahi1 KO mice. *p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001

Discussion

Our findings demonstrate that Ahi1 regulates ERβ protein expression by affecting GR nuclear translocation. Conversely, a reduction in Ahi1 expression decreases the cytoplasmic GR level, promotes ligand-dependent GR nuclear translocation and decreases the protein expression of ERβ in the nucleus; however, E2 can increase ERβ protein levels. Therefore, in female Ahi1 KO mice, high levels of E2 promoted ERβ protein expression and compensated for the Ahi1 deficiency-induced reduction in ERβ protein expression. Finally, these changes caused an increase in TPH2 and 5-HT levels, leading to antidepressant behavior. However, the lower E2 content in male mice could not compensate for the decrease in ERβ expression caused by Ahi1 KO, which led to a decrease in TPH2 and 5-HT levels and depressive behavior in male Ahi1 KO mice (Fig. 6D).

5-HT is a neurotransmitter that appears early in development and is widely distributed throughout the brain, and plays a central role in brain development, mood regulation, stress response, and risk of psychiatric disorders [1, 4, 7, 59]. Our previous findings suggested that 5-HT in the brain contributes to the depression-like behaviors induced by Ahi1 deficiency [12]. Our recent results indicated that Ahi1 inhibits GR nuclear translocation by directly interacting with GR [14]. More interestingly, the expression of ERβ decreased in the brains of male Ahi1 KO mice, and the reduction in expression was mainly in the nucleus, not the cytoplasm. Both GR and ER are ligand-responsive transcription factors that belong to the nuclear receptor superfamily [60]. Therefore, we speculated that Ahi1 might regulate ERβ transcription by interacting with GR. In Dex and/or GR-siRNA treated PC12 cells, luciferase reporter assays proved that Ahi1 inhibits GR nuclear translocation and regulates ERβ transcription; however, whether GR affects ERβ activity needs further investigation. The protein levels of 5-HT transporter SERT and the ERα were not changed in male Ahi1 KO and the mouse model of chronic restraint stress-induced depression-like behaviors; therefore, they may not be responsible for the decline in 5-HT levels and depression-like behavior in depressed mice. The ERβ/TPH2/5-HT pathway was inhibited in male Ahi1 KO mice and stressed mice, suggesting that the pathway has a critical role in regulating depressive behaviors. However, there was no change in the ERβ/TPH2/5-HT pathway or depression-like behavior in female Ahi1 KO and chronic restraint stressed mice, implying that there may be other signaling pathways involved in the regulation of 5-HT and behavior in female Ahi1 KO and chronic restraint stress mice.

Accumulating evidence suggests that there are sex differences in depression symptoms and the response to antidepressant treatment [33, 61]. It is clear that the prevalence of depression is higher in women than in men. In animals, male rats and mice seem to be more susceptible than females to depression-like effects [62, 63]. Sex differences are related to many factors, including artifacts and social or biological factors [64]. Our study used male and female Ahi1 KO and a mouse model of chronic restraint stress-induced depression-like behaviors to investigate the mechanisms underlying the decrease in 5-HT content-induced depression-like behaviors and sex differences, explaining the causes of sex differences from a biological perspective.

Estrogen has been used successfully to treat depression or depression-like behaviors in humans and rodents [15, 18, 65, 66]. Interestingly, E2-treated spayed macaques exhibited increased TPH mRNA expression in the dorsal raphe nucleus, indicating that E2 induces TPH gene expression in nonhuman primates [67]. In our study, the content of endogenous brain E2 was significantly higher in female mice than in male control mice, and the brain content of E2 was significantly decreased in male Ahi1 KO mice compared with male control mice; however, there was no difference between female Ahi1 KO mice and female control mice (Fig. 6A, B). Interestingly, E2 levels in plasma were not different between male Ahi1 KO mice and control mice (Fig. 6C). Therefore, brain E2 may play an antidepressant role by influencing TPH gene expression, leading to sex differences in depressive-like mouse models. These results suggested that E2 is involved in the regulation of depressive behaviors through the ERβ/TPH2/5-HT pathway and that the E2 level in the brain may be an important biochemical marker for the diagnosis of depression.

Conclusions

Ahi1 modulates the ERβ/TPH2/5-HT pathway through modulating GR nuclear translocation. The ERβ/TPH2/5-HT pathway is inhibited in male Ahi1 KO and chronic restraint stress-induced depressed mice but not in female mice. The difference in endogenous brain estrogen levels causes sex differences in depressive behaviors. Therefore, Ahi1 regulates serotonin production by the GR/ERβ/TPH2 pathway involving sex differences in depression-like behaviors; our findings may facilitate finding personalized treatments for patients with MDD.

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Abbreviations

5-HT:

5-Hydroxytryptamine

E2:

17β-Estradiol

ERβ:

Estrogen receptor-β

FST:

Forced swimming test

GR:

Glucocorticoid receptor

HPLC:

High-performance liquid chromatography

KO:

Knockout

MDD:

Major depressive disorders

PBS:

Phosphate-buffered saline

TPH2:

Tryptophan hydroxylase 2

TST:

Tail suspension test

References

  1. Gaspar P, Cases O, Maroteaux L. The developmental role of serotonin: news from mouse molecular genetics. Nat Rev Neurosci. 2003;4:1002–12.

    Article  CAS  PubMed  Google Scholar 

  2. Meneses A. 5-HT system and cognition. Neurosci Biobehav Rev. 1999;23:1111–25.

    Article  CAS  PubMed  Google Scholar 

  3. Muraki Y, Yamanaka A, Tsujino N, Kilduff TS, Goto K, Sakurai T. Serotonergic regulation of the orexin/hypocretin neurons through the 5-HT1A receptor. J Neurosci. 2004;24:7159–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Fossat P, Bacque-Cazenave J, De Deurwaerdere P, Cattaert D, Delbecque JP. Serotonin, but not dopamine, controls the stress response and anxiety-like behavior in the crayfish procambarus clarkii. J Exp Biol. 2015;218:2745–52.

    PubMed  Google Scholar 

  5. Dominguez-Lopez S, Howell R, Gobbi G. Characterization of serotonin neurotransmission in knockout mice: implications for major depression. Rev Neurosci. 2012;23:429–43.

    Article  CAS  PubMed  Google Scholar 

  6. Maurer-Spurej E, Pittendreigh C, Misri S. Platelet serotonin levels support depression scores for women with postpartum depression. J Psychiatry Neurosci. 2007;32:23–9.

    PubMed  PubMed Central  Google Scholar 

  7. Albert PR, Benkelfat C. The neurobiology of depression --revisiting the serotonin hypothesis. II. Genetic epigenetic and clinical studies. Philos Trans R Soc Lond B Biol Sci. 2013;368:20120535.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  8. Zhang X, Beaulieu JM, Sotnikova TD, Gainetdinov RR, Caron MG. Tryptophan hydroxylase-2 controls brain serotonin synthesis. Science. 2004;305:217.

    Article  CAS  PubMed  Google Scholar 

  9. Lopez de Lara C, Brezo J, Rouleau G, Lesage A, Dumont M, Alda M, Benkelfat C, Turecki G. Effect of tryptophan hydroxylase-2 gene variants on suicide risk in major depression. Biol Psychiatry. 2007;62:72–80.

    Article  CAS  PubMed  Google Scholar 

  10. Gutknecht L, Popp S, Waider J, Sommerlandt FM, Goppner C, Post A, Reif A, van den Hove D, Strekalova T, Schmitt A, et al. Interaction of brain 5-HT synthesis deficiency, chronic stress and sex differentially impact emotional behavior in Tph2 knockout mice. Psychopharmacology. 2015;232:2429–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Hiroi R, Handa RJ. Estrogen receptor-beta regulates human tryptophan hydroxylase-2 through an estrogen response element in the 5’ untranslated region. J Neurochem. 2013;127:487–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Ren L, Qian X, Zhai L, Sun M, Miao Z, Li J, Xu X. Loss of Ahi1 impairs neurotransmitter release and causes depressive behaviors in mice. PLoS ONE. 2014;9: e93640.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Oakley RH, Cidlowski JA. The biology of the glucocorticoid receptor: new signaling mechanisms in health and disease. J Allergy Clin Immunol. 2013;132:1033–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Wang B, Xin N, Qian X, Zhai L, Miao Z, Yang Y, Li S, Sun M, Xu X, Li XJ. Ahi1 regulates the nuclear translocation of glucocorticoid receptor to modulate stress response. Transl Psychiatry. 2021;11:188.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Rasgon NL, Dunkin J, Fairbanks L, Altshuler LL, Troung C, Elman S, Wroolie TE, Brunhuber MV, Rapkin A. Estrogen and response to sertraline in postmenopausal women with major depressive disorder: a pilot study. J Psychiatr Res. 2007;41:338–3.

    Article  PubMed  Google Scholar 

  16. Saletu B, Brandstatter N, Metka M, Stamenkovic M, Anderer P, Semlitsch HV, Heytmanek G, Huber J, Grunberger J, Linzmayer L, et al. Hormonal, syndromal and EEG mapping studies in menopausal syndrome patients with and without depression as compared with controls. Maturitas. 1996;23:91–105.

    Article  CAS  PubMed  Google Scholar 

  17. Xu Y, Sheng H, Bao Q, Wang Y, Lu J, Ni X. NLRP3 inflammasome activation mediates estrogen deficiency-induced depression- and anxiety-like behavior and hippocampal inflammation in mice. Brain Behav Immun. 2016;56:175–86.

    Article  CAS  PubMed  Google Scholar 

  18. Kiss A, Delattre AM, Pereira SI, Carolino RG, Szawka RE, Anselmo-Franci JA, Zanata SM, Ferraz AC. 17beta-estradiol replacement in young, adult and middle-aged female ovariectomized rats promotes improvement of spatial reference memory and an antidepressant effect and alters monoamines and BDNF levels in memory- and depression-related brain areas. Behav Brain Res. 2012;227:100–8.

    Article  CAS  PubMed  Google Scholar 

  19. Yang F, Tao J, Xu L, Zhao N, Chen J, Chen W, Zhu Y, Qiu J. Estradiol decreases rat depressive behavior by estrogen receptor beta but not alpha: no correlation with plasma corticosterone. NeuroReport. 2014;25:100–4.

    Article  CAS  PubMed  Google Scholar 

  20. Hughes ZA, Liu F, Platt BJ, Dwyer JM, Pulicicchio CM, Zhang G, Schechter LE, Rosenzweig-Lipson S, Day M. WAY-200070, a selective agonist of estrogen receptor beta as a potential novel anxiolytic/antidepressant agent. Neuropharmacology. 2008;54:1136–42.

    Article  CAS  PubMed  Google Scholar 

  21. Walf AA, Frye CA. Administration of estrogen receptor beta-specific selective estrogen receptor modulators to the hippocampus decrease anxiety and depressive behavior of ovariectomized rats. Pharmacol Biochem Behav. 2007;86:407–14.

    Article  CAS  PubMed  Google Scholar 

  22. Rocha BA, Fleischer R, Schaeffer JM, Rohrer SP, Hickey GJ. 17 Beta-estradiol-induced antidepressant-like effect in the forced swim test is absent in estrogen receptor-beta knockout (BERKO) mice. Psychopharmacology. 2005;179:637–43.

    Article  CAS  PubMed  Google Scholar 

  23. Fink G, Sumner BE, Rosie R, Grace O, Quinn JP. Estrogen control of central neurotransmission: effect on mood, mental state, and memory. Cell Mol Neurobiol. 1996;16:325–44.

    Article  CAS  PubMed  Google Scholar 

  24. Pandaranandaka J, Poonyachoti S, Kalandakanond-Thongsong S. Anxiolytic property of estrogen related to the changes of the monoamine levels in various brain regions of ovariectomized rats. Physiol Behav. 2006;87:828–35.

    Article  CAS  PubMed  Google Scholar 

  25. Calakos KC, Bhatt S, Foster DW, Cosgrove KP. Mechanisms underlying sex differences in cannabis use. Curr Addict Rep. 2017;4:439–53.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Karpyak VM, Geske JR, Hall-Flavin DK, Loukianova LL, Schneekloth TD, Skime MK, Seppala M, Dawson G, Frye MA, Choi DS, Biernacka JM. Sex-specific association of depressive disorder and transient emotional states with alcohol consumption in male and female alcoholics. Drug Alcohol Depend. 2019;196:31–9.

    Article  PubMed  Google Scholar 

  27. Schuch JJ, Roest AM, Nolen WA, Penninx BW, de Jonge P. Gender differences in major depressive disorder: results from the Netherlands study of depression and anxiety. J Affect Disord. 2014;156:156–63.

    Article  PubMed  Google Scholar 

  28. Scholl JL, Afzal A, Fox LC, Watt MJ, Forster GL. Sex differences in anxiety-like behaviors in rats. Physiol Behav. 2019;211:112670.

    Article  CAS  PubMed  Google Scholar 

  29. Rainville JR, Tsyglakova M, Hodes GE. Deciphering sex differences in the immune system and depression. Front Neuroendocrinol. 2018;50:67–90.

    Article  CAS  PubMed  Google Scholar 

  30. Ferrari AJ, Somerville AJ, Baxter AJ, Norman R, Patten SB, Vos T, Whiteford HA. Global variation in the prevalence and incidence of major depressive disorder: a systematic review of the epidemiological literature. Psychol Med. 2013;43:471–81.

    Article  CAS  PubMed  Google Scholar 

  31. Marcus SM, Kerber KB, Rush AJ, Wisniewski SR, Nierenberg A, Balasubramani GK, Ritz L, Kornstein S, Young EA, Trivedi MH. Sex differences in depression symptoms in treatment-seeking adults: confirmatory analyses from the sequenced treatment alternatives to relieve depression study. Compr Psychiatry. 2008;49:238–46.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Kornstein SG, Pedersen RD, Holland PJ, Nemeroff CB, Rothschild AJ, Thase ME, Trivedi MH, Ninan PT, Keller MB. Influence of sex and menopausal status on response, remission, and recurrence in patients with recurrent major depressive disorder treated with venlafaxine extended release or fluoxetine: analysis of data from the PREVENT study. J Clin Psychiatry. 2014;75:62–8.

    Article  PubMed  Google Scholar 

  33. Sramek JJ, Murphy MF, Cutler NR. Sex differences in the psychopharmacological treatment of depression. Dialogues Clin Neurosci. 2016;18:447–57.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Marcus SM, Young EA, Kerber KB, Kornstein S, Farabaugh AH, Mitchell J, Wisniewski SR, Balasubramani GK, Trivedi MH, Rush AJ. Gender differences in depression: findings from the STAR*D study. J Affect Disord. 2005;87:141–50.

    Article  PubMed  Google Scholar 

  35. Liu X, Sun YX, Zhang CC, Zhang XQ, Zhang Y, Wang T, Ma YN, Wang H, Su YA, Li JT, Si TM. Vortioxetine attenuates the effects of early-life stress on depression-like behaviors and monoamine transporters in female mice. Neuropharmacology. 2021;186:108468.

    Article  CAS  PubMed  Google Scholar 

  36. Hankin BL, Abramson LY, Moffitt TE, Silva PA, McGee R, Angell KE. Development of depression from preadolescence to young adulthood: emerging gender differences in a 10-year longitudinal study. J Abnorm Psychol. 1998;107:128–40.

    Article  CAS  PubMed  Google Scholar 

  37. Pitychoutis PM, Nakamura K, Tsonis PA, Papadopoulou-Daifoti Z. Neurochemical and behavioral alterations in an inflammatory model of depression: sex differences exposed. Neuroscience. 2009;159:1216–32.

    Article  CAS  PubMed  Google Scholar 

  38. Goswami DB, May WL, Stockmeier CA, Austin MC. Transcriptional expression of serotonergic regulators in laser-captured microdissected dorsal raphe neurons of subjects with major depressive disorder: sex-specific differences. J Neurochem. 2010;112:397–409.

    Article  CAS  PubMed  Google Scholar 

  39. Nishizawa S, Benkelfat C, Young SN, Leyton M, Mzengeza S, de Montigny C, Blier P, Diksic M. Differences between males and females in rates of serotonin synthesis in human brain. Proc Natl Acad Sci U S A. 1997;94:5308–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Jovanovic H, Lundberg J, Karlsson P, Cerin A, Saijo T, Varrone A, Halldin C, Nordstrom AL. Sex differences in the serotonin 1A receptor and serotonin transporter binding in the human brain measured by PET. Neuroimage. 2008;39:1408–19.

    Article  PubMed  Google Scholar 

  41. Binder EB, Kunzel HE, Nickel T, Kern N, Pfennig A, Majer M, Uhr M, Ising M, Holsboer F. HPA-axis regulation at in-patient admission is associated with antidepressant therapy outcome in male but not in female depressed patients. Psychoneuroendocrinology. 2009;34:99–109.

    Article  CAS  PubMed  Google Scholar 

  42. DeSantis SM, Baker NL, Back SE, Spratt E, Ciolino JD, Moran-Santa Maria M, Dipankar B, Brady KT. Gender differences in the effect of early life trauma on hypothalamic-pituitary-adrenal axis functioning. Depress Anxiety. 2011;28:383–92.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Back SE, Waldrop AE, Saladin ME, Yeatts SD, Simpson A, McRae AL, Upadhyaya HP, Contini Sisson R, Spratt EG, Allen J, et al. Effects of gender and cigarette smoking on reactivity to psychological and pharmacological stress provocation. Psychoneuroendocrinology. 2008;33:560–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Eid RS, Gobinath AR, Galea LAM. Sex differences in depression: insights from clinical and preclinical studies. Prog Neurobiol. 2019;176:86–102.

    Article  PubMed  Google Scholar 

  45. Porcelli S, Pae CU, Han C, Lee SJ, Patkar AA, Masand PS, Balzarro B, Alberti S, De Ronchi D, Serretti A. Abelson helper integration site-1 gene variants on major depressive disorder and bipolar disorder. Psychiatry Investig. 2014;11:481–6.

    Article  PubMed  PubMed Central  Google Scholar 

  46. Weng L, Lin YF, Li AL, Wang CE, Yan S, Sun M, Gaertig MA, Mitha N, Kosaka J, Wakabayashi T, et al. Loss of Ahi1 affects early development by impairing BM88/Cend1-mediated neuronal differentiation. J Neurosci. 2013;33:8172–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Zhang G, Chen L, Yang L, Hua X, Zhou B, Miao Z, Li J, Hu H, Namaka M, Kong J, Xu X. Combined use of spatial restraint stress and middle cerebral artery occlusion is a novel model of post-stroke depression in mice. Sci Rep. 2015;5:16751.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Cheng Y, Sun M, Chen L, Li Y, Lin L, Yao B, Li Z, Wang Z, Chen J, Miao Z, et al. Ten-eleven translocation proteins modulate the response to environmental stress in mice. Cell Rep. 2018;25(3194–3203): e3194.

    Article  CAS  Google Scholar 

  49. Wang B, Zheng Y, Shi H, Du X, Zhang Y, Wei B, Luo M, Wang H, Wu X, Hua X, et al. Zfp462 deficiency causes anxiety-like behaviors with excessive self-grooming in mice. Genes Brain Behav. 2017;16:296–307.

    Article  CAS  PubMed  Google Scholar 

  50. Miao Z, He Y, Xin N, Sun M, Chen L, Lin L, Li J, Kong J, Jin P, Xu X. Altering 5-hydroxymethylcytosine modification impacts ischemic brain injury. Hum Mol Genet. 2015;24:5855–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Wang B, Zhang Y, Dong H, Gong S, Wei B, Luo M, Wang H, Wu X, Liu W, Xu X, et al. Loss of Tctn3 causes neuronal apoptosis and neural tube defects in mice. Cell Death Dis. 2018;9:520.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Xu X, Yang H, Lin YF, Li X, Cape A, Ressler KJ, Li S, Li XJ. Neuronal Abelson helper integration site-1 (Ahi1) deficiency in mice alters TrkB signaling with a depressive phenotype. Proc Natl Acad Sci U S A. 2010;107:19126–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Le Grand A, Andre-Leroux G, Marteil G, Duval H, Sire O, Le Tilly V. Investigating the in vitro thermal stability and conformational flexibility of estrogen receptors as potential key factors of their in vivo activity. Biochemistry. 2015;54:3890–900.

    Article  PubMed  CAS  Google Scholar 

  54. Sharma HR, Thakur MK. Correlation of ERalpha/ERbeta expression with dendritic and behavioural changes in CUMS mice. Physiol Behav. 2015;145:71–83.

    Article  CAS  PubMed  Google Scholar 

  55. Doering JE, Kane K, Hsiao YC, Yao C, Shi B, Slowik AD, Dhagat B, Scott DD, Ault JG, Page-McCaw PS, Ferland RJ. Species differences in the expression of Ahi1, a protein implicated in the neurodevelopmental disorder Joubert syndrome, with preferential accumulation to stigmoid bodies. J Comp Neurol. 2008;511:238–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Sheng G, Xu X, Lin YF, Wang CE, Rong J, Cheng D, Peng J, Jiang X, Li SH, Li XJ. Huntingtin-associated protein 1 interacts with Ahi1 to regulate cerebellar and brainstem development in mice. J Clin Invest. 2008;118:2785–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Weiser MJ, Wu TJ, Handa RJ. Estrogen receptor-beta agonist diarylpropionitrile: biological activities of R- and S-enantiomers on behavior and hormonal response to stress. Endocrinology. 2009;150:1817–25.

    Article  CAS  PubMed  Google Scholar 

  58. Perry LM, Goldstein-Piekarski AN, Williams LM. Sex differences modulating serotonergic polymorphisms implicated in the mechanistic pathways of risk for depression and related disorders. J Neurosci Res. 2017;95:737–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Brummelte S, Mc Glanaghy E, Bonnin A, Oberlander TF. Developmental changes in serotonin signaling: Implications for early brain function, behavior and adaptation. Neuroscience. 2017;342:212–31.

    Article  CAS  PubMed  Google Scholar 

  60. Evans RM. The steroid and thyroid hormone receptor superfamily. Science. 1988;240:889–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Young EA, Kornstein SG, Marcus SM, Harvey AT, Warden D, Wisniewski SR, Balasubramani GK, Fava M, Trivedi MH, John Rush A. Sex differences in response to citalopram: a STAR*D report. J Psychiatr Res. 2009;43:503–11.

    Article  PubMed  Google Scholar 

  62. Brotto LA, Barr AM, Gorzalka BB. Sex differences in forced-swim and open-field test behaviours after chronic administration of melatonin. Eur J Pharmacol. 2000;402:87–93.

    Article  CAS  PubMed  Google Scholar 

  63. Palanza P. Animal models of anxiety and depression: how are females different? Neurosci Biobehav Rev. 2001;25:219–33.

    Article  CAS  PubMed  Google Scholar 

  64. Frackiewicz EJ, Sramek JJ, Cutler NR. Gender differences in depression and antidepressant pharmacokinetics and adverse events. Ann Pharmacother. 2000;34:80–8.

    Article  CAS  PubMed  Google Scholar 

  65. Dennis CL, Ross LE, Herxheimer A. Oestrogens and progestins for preventing and treating postpartum depression. Cochrane Database Syst Rev. 2008. https://doi.org/10.1002/14651858.CD001690.pub2.

    Article  PubMed  PubMed Central  Google Scholar 

  66. Studd J, Panay N. Are oestrogens useful for the treatment of depression in women? Best Pract Res Clin Obstet Gynaecol. 2009;23:63–71.

    Article  PubMed  Google Scholar 

  67. Pecins-Thompson M, Brown NA, Kohama SG, Bethea CL. Ovarian steroid regulation of tryptophan hydroxylase mRNA expression in rhesus macaques. J Neurosci. 1996;16:7021–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by the grants from the National Science Foundation of China (81120108011, 81771454, 81974244, 82071511 and 82101793), the National Key R&D Program of China (2019YFA0802600, 2017YFE0103700), the Priority Academic Program Development of Jiangsu Higher Education Institutions of China and the Natural Science Foundation of Jiangsu Province (BK20210092) and Suzhou Natural Science foundation (SKJY2021061). Jan-Ake Gustafsson is grateful to the Robert A. Welch Foundation for a grant (E-30007). We appreciate the support of Jiangsu Innovative and Enterpreneurial Talent Programme (JSSCBS20211567) and Suzhou Gusu health talent program.

Author information

Authors and Affiliations

Authors

Contributions

XX designed and supervised the study. BW, HS, and ZM performed the experiments. HS, BW, LR, ZM, BW, HY, and MS analyzed the data. BW, HS, MS, and X.X. wrote the manuscript. All authors discussed the results and commented on the manuscript. XF, JG modify the article language. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Miao Sun or Xingshun Xu.

Ethics declarations

Ethics approval and consent to participate

All animal experiments were performed in accordance with the guidelines of the University Committee on Animal Care of Soochow University.

Consent for publication

All authors have approved for publication.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1: Table S1.

Primers used in this study.

Additional file 2: Fig. S1.

There were sex differences in ERβ/TPH2/5-HT pathway and depression-like behaviors in the brainstem of Ahi1 KO mice. A After female and male Ahi1 KO mice were sacrificed, brainstem tissues were collected, and TPH2 protein expression was examined by Western blot analysis. N=3. B TPH2 mRNA levels were quantified by quantitative PCR in the brainstem tissues of female and male Ahi1 KO mice. N=3. C ERβ mRNA expression in brainstem tissue of female and male Ahi1 KO mice was examined by quantitative PCR. N=3-5. D ERβ protein expression in brainstem tissue of female and male Ahi1 KO mice was examined by Western blot analysis. N=3. E ERα protein expression was examined in hippocampus tissue of male Ahi1 KO mice by Western blot analysis. N=3. F Quantity analysis of ERβ expression fluorescence intensity and the number of ERβ-positive cells in the brainstem in male Ahi1 KO mice. *p<0.05, **p<0.01, ***p<0.001 versus Control.

Additional file 3: Fig. S2.

Stress led to depression-like behaviors and a decrease of Ahi1 in the hippocampus of male mice, but not in female mice. A, B After the healthy female and male C57 mice were stressed for 2 weeks, the behavioral tests were performed in the stressed mice. the immobility time in TST and FST tests was recorded. N=8-11. C, D After female and male C57 mice were stressed for 2 weeks, the hippocampal tissues of male and female stressed mice were collected and their Ahi1 content was detected by Western blot. N=4. **p<0.01, ***p<0.001 versus Control).

Additional file 4: Fig. S3.

There were sex differences in ERβ/TPH2/5-HT pathway in stressed mice. A, B After female and male stressed mice were sacrificed, hippocampus tissue was collected, TPH2 mRNA levels were quantified by quantitative PCR (A) and TPH2 protein expression was examined by Western blot analysis (B). N=3. C ERβ mRNA levels were quantified by quantitative PCR in the hippocampus tissue from female and male stressed mice. N=3-4. D ERβ protein expression was performed in hippocampus tissue from female and male stressed mice. N=3. E SERT protein expression was examined in the hippocampus tissue of male stressed mice by Western blot analysis. N=3. **p<0.01 versus Control.

Additional file 5: Fig. S4.

Dexamethasone promotes GR nuclear translocation and inhibited the transcription of ERβ mRNA. ERβ and GR protein expression were detected in the nucleus (A) and cytoplasm (B) in Dex-treated PC12 cells for 72 h by Western blot analysis. N=3. **p<0.01, ***p<0.001 versus Control.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, B., Shi, H., Ren, L. et al. Ahi1 regulates serotonin production by the GR/ERβ/TPH2 pathway involving sexual differences in depressive behaviors. Cell Commun Signal 20, 74 (2022). https://doi.org/10.1186/s12964-022-00894-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12964-022-00894-4

Keywords