Materials and methods
Patients and healthy controls
Our research included 17 healthy controls and 35 patients with a first episode of MDD (figure 1). Enrolment was open to adult men and women aged 18–65 years old. Eligible subjects required a Diagnostic and Statistical Manual of Mental Disorders Structured Clinical Interview (Fourth Edition) diagnosis of MDD7; screening was conducted by psychiatrists from Nanjing Brain Hospital of Nanjing Medical University. The study started in June 2018 and concluded in December 2021. HAMD (24-item version) was used to evaluate the severity of depressive symptoms, such as depression, loss of interest, decreased energy, fatigue and pain8; and the Hamilton Rating Scale for Anxiety (HAMA; 17-item version) was used to evaluate the severity of anxiety symptoms.9 Patients with MDD were identified based on a score of 20 or higher on the HAMD, with the assessment conducted by an independent, trained rater. The exclusion criteria were as follows: any antidepressant treatment for more than a week, history of brain trauma, severe personality disorder, risk of suicide, pregnancy, current psychotherapy of any kind and diagnosis of bipolar disorder (figure 1). Information on enrolment is provided in online supplemental table S1,S2. The blood sample (5 mL) of patients with MDD was collected at the same time (07:00–08:00; the second day after completing the HAMD and HAMA scores) in the inpatient ward with BD Vacutainer tubes and centrifuged at 3000 g for 10 min at 4°C within 2 hours. The blood sample of the healthy control participants, who had no history of mental, neurological or serious physical diseases, and who volunteered to take part in this research was collected at the same time (07:00–08:00) in the Nanjing Brain Hospital of Nanjing Medical University. All participants’ evaluation and blood sample collection were completed within 2 months. The serum was then aliquoted from the supernatant and stored at −80°C before further processing.
Figure 1Flowchart of the enrolment and follow-up of subjects of the study. 17 healthy controls and 35 patients with first episode of MDD were enrolled according to the inclusion and exclusion criteria. Patients with MDD were identified based on a HAMD score of 20 or higher. HAMD, Hamilton Depression Scale; MDD, major depressive disorder.
Animals
Eight-week-old male C57BL/6 mice (Animal Core Facility of Nanjing Medical University) were used for establishing the CRS model. Mice were maintained at a constant room temperature (18°C–22°C), with a controlled illumination (12:12 hours light/dark cycle), relative humidity of 30%–50%, and with food and water available ad libitum.
Establishment of the CRS mouse model
The CRS was established by restraining the mice for 5 hours per day (09:00–14:00) for 4 consecutive weeks, as previously described.10 During CRS, the mice were kept in isolation and placed horizontally in plastic centrifuge tubes (50 mL; 3 cm in diameter and 11.5 cm in height) with 10 pinholes with a diameter of 0.5 cm on the surface of each tube for ventilation. Toilet paper was placed at the cap site to absorb mouse faeces to minimise discomfort. After the constriction was released, the mice were put back in their home cages and given ad libitum access to food and water. The control mice were left undisturbed in their home cages. The mice were protected from any physical wounding during the modelling period.
Behavioural testing
After the modelling was completed, the mice received behavioural tests to analyse the levels of depression and anxiety in the Small Animal Behaviour Testing Laboratory of Animal Core Facility of Nanjing Medical University. Behavioural data were collected by two individuals who were unaware of animal group allocations, minimising potential experimenter-induced bias. The behavioural tests were conducted in the following order: sucrose preference test (SPT), open field test (OFT), social interaction test, elevated plus maze (EPM) test, forced swimming test (FST) and tail suspension test (online supplemental materials), for these behavioural tests gradually increased the stress in mice. Each behavioural test was conducted each day in the above order to minimise stress stimuli and potential impacts on test results. All tests were performed by experimenters who were blind to the treatment schedule.
CRS grouping
The CRS mice with a lower sugar preference rate and longer swimming immobility time than the average of the control group were categorised as belonging to the CRS susceptibility group, and those with a higher sugar preference rate and shorter swimming immobility time than the average level of the control group were termed CRS-resistant mice. Afterwards, five mice in each group were randomly selected to collect fresh brain tissue for western blot and enzyme-linked immunosorbent assay (ELISA). An additional five brain samples per group underwent Golgi silver staining to detect morphological changes in the cortical neurons. The mice that did not meet the above classification criteria were excluded from the biochemical and pathological analyses.
Viral construction and stereotaxic surgery
For overexpression of miR-451a in vivo, viral constructs were designed using adeno-associated virus serotype 9 (AAV9) vectors (Genechem, China), encoding a pri-mmu-miR-451a-GFP fusion protein (Ad_OE-miR-451a) or green fluorescent protein (GFP) only as a negative control, under the control of the cytomegalovirus (CMV) promoter (Ad_OE-scramble).
The mice were deeply anaesthetised with sodium pentobarbital (50 mg/kg, intraperitoneal injection) and then placed in a stereotaxic apparatus. Bilateral microinfusions were made using a 1 µL Hamilton microsyringe with a stainless-steel infusion needle (33-gauge) into the mouse mPFC at the following coordinates: anterior-posterior: +1.90 mm; medial-lateral: ±0.5 mm; and dorsal-ventral: −2.3 mm relative to the bregma. For miR-451a overexpression experiments, a total volume of 0.25 µL of AAV-miR-451a-OE (2.0 E+13 genome copies per millilitre) or control virus was injected into the mPFC of each hemisphere over an 8 min period. The infusion needles were left inside the mPFC area during a 5 min pause to limit virus reflux. The mice recovered for 14 days before initiating the CRS procedure.
Sample and serum preparation
On the next morning after the final behavioural tests, the mice were anaesthetised and their blood was collected via the ophthalmic artery. Blood samples were allowed to coagulate in centrifuge tubes at room temperature for 30 min and subsequently centrifuged at 1500 g for 10 min. The serum was separated and stored at −80°C until the biochemical estimations were carried out. The mice were perfused transcardially with 50 mL of 0.9% saline right after blood collection, and the mPFC was quickly extracted from the whole brain, frozen by liquid nitrogen, then stored at −80°C until use.
RNA extraction and analysis
miRNA and mRNA levels were determined using real-time quantitative polymerase chain reaction (qPCR). Total RNA was isolated from human serum or from the mPFC and serum of mice using RNAiso Plus (Takara) according to the manufacturer’s protocol, with minor modifications, and quantified by NanoDrop 2000 (Thermofisher, USA). Human serum miRNA detection was carried out in 384-well plates using the Q7 real-time PCR detection system (Thermofisher), and the small nucleolar RNA RNU6B was used as endogenous control. The expression of miRNAs was relativised to (ie, normalised to) the healthy control group and presented as fold change. For mouse serum miRNA analysis, the Caenorhabditis elegans-specific miRNA, cel-miR-39, was added to mouse serum as exogenous control. miRNAs were reverse-transcribed using a poly (A) tailing reaction and a universal cDNA synthesis kit (Sangon Biotech, China), and then qPCR was conducted using the SYBR Green MicroRNA qPCR Kit (B532461; Sangon Biotech), with the primers listed in online supplemental table S3.
Reverse transcription PCR for mRNA was carried out using a Takara PrimeScript RT Reagent Kit (no: RR036A), and mRNA expression was evaluated using a QuantiTect SYBR Green PCR Kit (RR820A; Takara, Japan) with an ABI 7300 Fast Real-Time PCR System (Applied Biosystems, Foster City, California) by real-time PCR. The results were analysed and presented relative to threshold cycle values normalised to the expression of glyceraldehyde-3-phosphate dehydrogenase (GAPDH), then converted to fold changes. The sequences of primers for real-time PCR analysis are listed in online supplemental table S4.
Western blot
Western blot analysis was performed following standard protocols. Briefly, homogenised protein of the mPFC was extracted from samples using RIPA lysis buffer (Beyotime, China) with phosphatase inhibitors (Roche, Switzerland), and concentration was determined by the Bradford assay (Beyotime). The analysis of protein was performed according to standard Sodium Dodecy Sulfate Polyacrylamide Gel Electrophoresis (SDS-PAGE). Protein was separated by 8%–15% SDS-PAGE and electrophoretically transferred onto polyvinylidene fluoride (PVDF) membranes (Millipore, USA). After blocking for 1 hour in 5% skimmed milk in tris-buffered saline with Tween 20 (TBST), the transferred membranes were incubated with primary anti-mouse antibodies (online supplemental table S5) at 4°C overnight, and then with horseradish peroxidase (HRP)-conjugated secondary antibody for 1 hour at room temperature. The blots were visualised by an enhanced chemiluminescence (ECL) kit (Tanon, China). Values for protein levels were calculated using ImageJ (National Institutes of Health, USA) and normalised to GAPDH. Fold changes against values in the control group were calculated and used to perform statistical analysis. At least three mice in each group were used for western blot analysis.
Enzyme-linked immunosorbent assay
Quantitative determination of serum corticosterone (CORT) was performed using a commercially available ELISA kit (R&D Systems, USA) with high sensitivity (0.047 ng/mL) according to the manufacturer’s instructions.
Golgi staining and dendritic spine analysis
Golgi staining was performed using the FD Rapid GolgiStain Kit (FD Neuro Technologies, USA). The dendritic spines of the neurons in layers 3–5 of mPFC were imaged using a 100× oil objective on a digital camera (Leica Microsystems, Germany). The dendritic spines on secondary or tertiary dendritic branches were analysed, and each branch was at least 10 µm in length. Five mPFC neurons per section and three sections per mouse were analysed.
FISH and immunofluorescence
For immunofluorescence, the brain sections at 15 µm containing the mPFC were blocked at room temperature for 1 hour in 5% bovine serum albumin with 0.3% phosphate-buffered saline with Tween 20 (PBST), incubated with rabbit anti-corticotropin-releasing factor receptor 1 (CRFR1) (1:600; SAB, 40785) at 4°C overnight. After phosphate buffered saline (PBS) washing, sections were incubated for 2 hours at room temperature with Alexa Fluor 555 goat anti-rabbit IgG (H+L) highly cross-adsorbed secondary antibody (1:1000; Invitrogen, 31572), then incubated for 5 min at room temperature in 1.5 µM DAPI (4′,6-diamidino-2-phenylindole; Invitrogen, D1306) and sealed with buffered PBS/glycerol.
Locked-nucleotide modified and Cy3-labelled miR-451a probes were designed and synthesised by Genepharma (China); the probe sequence was AAC+TCAGTAA+TGGTAACGGT+TT; the ‘+’ sign was the modification site of the locked nucleotide. The probe signals were detected with a fluorescence in situ hybridisation (FISH) kit (RiboBio, China) according to the manufacturer’s instructions. Briefly, 15 μm thick brain sections were incubated with miR-451a probes for 10 hours at 37°C after treatment with prehybridisation solution. After washing with PBS, some FISH sections were performed following the immunofluorescent procedure as described above. The primary antibodies included rabbit antineuronal nuclear antigen (NeuN) (1:500; Abcam, ab177487), mouse anti-glial fibrillar acidic protein (GFAP) (1:800; Millipore, MAB360) and rabbit anti-GFAP (1:400; Abcam, ab7260); the secondary antibodies were Alexa Fluor 488 donkey anti-mouse IgG (1:1000; Invitrogen, A21202) and Alexa Fluor 488 donkey anti-rabbit IgG (1:1000; Invitrogen, A21206).
The fluorescent images at 400× magnification were captured by a Zeiss LSM710 confocal microscope (Zeiss, Germany) with a constant exposure time, offset and gain for each fluorescent staining marker from the mPFC. The total fluorescence intensity of CRFR1 or Cy3-labelled miR-451a in the mPFC was quantified by ImageJ V.1.52a (National Institutes of Health) and shown as a ratio normalised to control. Three to four brain sections in each set were averaged for each mouse, and four to five mice were averaged for each group.
Cell culture and transfections
Human embryonic kidney cells (HEK293T) and mouse brain cells (Neuro-2a, N2A) were cultured in Dulbecco’s modified eagle’s medium supplemented with 10% fetal bovine serum (Gibco), 100 U/mL penicillin and 100 µg/mL streptomycin (Invitrogen, USA) at 37 °C with 5% CO2.
For miR-451a downregulation, an miR-451a antisense hairpin inhibitor (RiboBio) was used. Mature double-stranded miR-451a was designed as a mimic (RiboBio) to imitate the overexpression of miR-451a. For ATF2 knockdown, an siRNA duplex against mouse ATF2 or a scrambled negative control was used (Genepharma). Briefly, cells were seeded in 12-well plates at a density of 2×105 cells per well. Twenty-four hours after seeding, mimics (50 nM) or inhibitors (100 nM) of miRNA and negative control or ATF2 siRNA (100 nM) were transfected into cells by Lipofectamine 2000 (Invitrogen, USA) following the manufacturer’s instructions. Cell RNA and protein were obtained 48 hours after transfection.
Primary cortical neurons were dissociated from postnatal day-1 C57BL/6 J mice and cultured at 5×106 cells/mL in Neurobasal-A medium (Invitrogen, Carlsbad, California) supplemented with B27 (Invitrogen, USA) and glutamine (Sigma, USA) on substrates precoated with 0.5 mg/mL of poly-L-lysine (Sigma). On the ninth day, the neurons were transfected with miR-451a mimic (75 nM) or an equal amount of scramble sequence using INTERFER in transfection reagent (Polyplus Transfection, PT-409-10) according to the manufacturer’s protocol, and then were treated with 10 µm CORT (Selleck, USA) or vehicle (dimethyl sulfoxide (DMSO) at 1:1000 dilution) 6 hours later and continued to be cultured for an additional 3 days. The primary cortical neurons were labelled with the PKH26 red fluorescent cell lipid membrane marker (Sigma) to make the dendritic arbours visible. Three wells were replicated for each treatment, with five different fields of view taken randomly per well using a digital microscope (Zeiss), and spines on dendritic arbours that were at least 50 µm long were analysed.
Dual-luciferase assays
Normal and mutated 3′ untranslated region (UTR) sequences (Atf2 mmu-miR-451a wild type, 5′-GGAAGAATATTAGAAACACTTTTTTTTAAGTGAGTGAAAGTATGGTAAGACGGTTAGTGCTTTGTGCACTTCTTAGACTAATCAA-3′; Atf2 mmu-miR-451a mutant, 5′-GGAAGAATATTAGAAACACTTTTTTTTAAGTGAGTGAATCATACCAAATTGCAAAGTGCTTTGTGCACTTCTTAGACTAATCAA-3′) of ATF2 were subcloned into the pmirGLO Dual-Luciferase miRNA target expression vector (Tsingke, Nanjing, China). HEK293T cells were transfected with the transfection-ready luciferase reporter construct and the mimics and inhibitors of miR-451a and negative control (50 nM per well). Forty-eight hours after transfection, the cells were lysed and luciferase reporter activities were assayed using the Dual-Luciferase Reporter Assay System (E1910; Promega) according to the manufacturer’s protocol. We normalised firefly luciferase activity to Renilla luciferase activity for each sample.
Statistical analysis
Data represent mean±SEM and were analysed using GraphPad Prism V.8.0 software (GraphPad Software, USA). Statistical differences between the two groups were analysed using two-tailed unpaired Student’s t-test or Mann-Whitney U test. Normality was determined through Anderson-Darling test for normality. Differences between multiple groups were assessed by one-way or two-way analysis of variance, followed by Tukey’s multiple comparison test. The correlation between miR-451a and behavioural indexes was measured by Pearson’s correlation analysis. The correlation between miR-451a, miR-1202 in serum, and question 7 (Q7) and question 23 (Q23) in the HAMD of patients with MDD was measured by non-parametric Spearman correlation. P value at 0.05 was considered a statistically significant level.