Clinical samples
From November 2018 to November 2019, patients with PCOS (aged 22–36 years) diagnosed using the Rotterdam criteria [21] were randomly assigned (using computer-generated random numbers) to undergo treatment with GH (PCOS-GH) or without GH (PCOS-C). Written informed consent was obtained from each participant. The study also conforms to the Declaration of Helsinki for Medical Research involving Human Subjects (2013 revision). Age-matched women (aged 25–37 years) who underwent in vitro fertilization and embryo transfer (IVF-ET) because of tubal infertility were recruited as non-PCOS controls. Primary GCs were collected from PCOS-C (n = 31), PCOS-GH (n = 30), and non-PCOS groups (n = 32). Exclusion criteria included hydrosalpinx, systemic lupus erythematosus, or sicca syndrome; uncontrolled endocrinopathy such as diabetes, hyperthyroidism, hypothyroidism, and hyperprolactinemia; or currently taking anti-OS medicine such as vitamin E, vitamin C, and Coenzyme Q10. Medical history-related information such as menstrual cycle regularity, duration of infertility, and treatment was collected from all the participants. Physical examinations included measurements of height, body weight, waist circumference, and hip circumference. Body weight index (BMI) was calculated as weight divided by height squared (kg/m2). The waist-to-hip ratio (WHR) was calculated as the waist circumference divided by the hip circumference. Androgen-related symptoms of hirsutism and acne were evaluated as previously reported [22]. Antral follicle count (AFC) were assessed by performing transvaginal ultrasound examination on days 2–3 of menstruation or progestin-induced withdrawal bleeding.
Plasma glucose, estradiol (E2), progesterone, total testosterone (TT), luteinizing hormone (LH), follicle-stimulation hormone (FSH), sex hormone binding globulin (SHBG), and fasting insulin (FINS) levels were measured as reported previously [22]. The free androgen index (FAI) was calculated as TT (nmol/L)/SHBG (nmol/L) × 100. The homeostasis model assessment (HOMA-IR) index was calculated as fasting glucose (mmol/L) × fasting insulin (mIU/L)/22.5 [22]. The intra- and inter-assay coefficients of variation for these values were <5% and <10%, respectively.
All the patients underwent IVF-ET according to the gonadotropin-releasing hormone antagonist protocol. Recombined follicular stimulation hormone (rFSH) (Gonal-F; Merck-Serono KGaA., Darmstadt, Germany) was administered starting from day 2 of the menstrual cycle. The dose of rFSH was adjusted according to follicular growth. Other than rFSH, patients in the PCOS-GH group were subcutaneously administered with 4 IU/d of recombinant human GH (Jintropin, Changchun GeneScience Pharmaceutical Co., Ltd., Changchun, Jilin, China) until the trigger day. Cetrorelix (Cetrotide; Merck-Serono KGaA.) was administered when one of the criteria was met: serum E2 > 300 pg/mL, leading follicle diameter reached 13–14 mm, LH > 10 IU/L. Recombinant human chorionic gonadotropin (Ovitrelle®; Merck-Serono KGaA., Darmstadt, Germany) was administered as the trigger when the diameters of at least two follicles reached ≥ 18 mm. After 36 hours, oocytes were retrieved under transvaginal ultrasound guidance.
Primary GC isolation
After oocyte retrieval, follicular fluid (FF) was collected from follicles with a diameter ≥ 16 mm measured on the retrieval day and immediately separated by centrifugation at 700 ×g for 5 min at room temperature. The precipitates were suspended in left 2 ml of FF and gently layered into 3 mL of 50% lymphocyte separation medium (Solarbio Science and Technology Corporation, Beijing, China). After centrifugation at 700 ×g for 10 min at room temperature to remove red blood cells and debris, GCs layered at the interface of the gradient were collected and washed twice with 5 mL of phosphate-buffered saline (Nanjing KeyGen Biotech. Co., Ltd., Nanjing, Jiangsu, China). Furthermore, residual red blood cells were removed using red blood cell lysis buffer (Solarbio Science and Technology Corporation). GCs from each patient were collected separately and considered as one sample. Intracellular ROS levels, mitochondrial membrane potential (MMP) and apoptosis of a portion of GCs were examined immediately. The remaining GCs were stored at −80°C immediately for mRNA and protein detection.
Detection of intracellular ROS levels
ROS generation in GCs was estimated using 2′,7′-diclorodihydrofluorescein di-acetate (H2-DCFDA) method by ROS assay kit (Beyotime Biotechnology Co., Ltd., Shanghai, China). Briefly, GCs were resuspended in PBS and incubated with 10 µM H2-DCFDA in the dark for 25 min at 37°C and then incubated together with 10 µg/mL 4',6-diamidino-2-phenylindole (DAPI) (NeoFroxx, Frankfurt, Germany) for 5 min. After the cells were washed three times with PBS, GC suspensions were added to glass slides, and examined by fluorescence microscopy (Olympus Corporation, Tokyo, Japan). The examination wavelength was 488 nm, and the emission wavelength was 525 nm.
Similar with the above protocol but without DAPI, NanoDrop UV-Vis spectrophotometry (Thermo Scientific, MA, USA) was used to measure the intracellular ROS level in another part of GCs. The fluorescence intensities are calculated as the intensity of the PCOS group relative to that of the control group (non-PCOS group).
Apoptosis assay
Apoptosis of GCs were detected using the Annexin V-FITC apoptosis detection kits (KeyGEN Bio TECH Co., Ltd.). Briefly, 1×10 5/ Test of GCs were resuspended in 500 µL binding buffer, then labeled with Annexin Ⅴ-FITC (5 µL) and propidium iodide (PI) (5 µL) for 15 min in the dark at room temperature. After 1 hour, the green (Annexin V-FITC) and red (PI) fluorescence were examined by flow cytometry (MilliporeSigma Co., Ltd., Burlington, MA, USA). The examination wavelength was 488 nm, and the emission wavelength was 530 nm.
Detection of MMP
The MMP of GCs was examined using JC-1 Apoptosis Detection Kits (KeyGEN Bio TECH Co., Ltd.). In brief, 1×10 5/ Test of GCs were resuspended and incubated with 500 µL JC-1 reagent solution at 37˚C in the dark for 15 min. JC-1 accumulates in functional mitochondria with high ΔΨm and forms aggregates that emit red fluorescence. When mitochondrial transmembrane potential is depolarized with low ΔΨm, JC-1 releases from the mitochondria and forms monomers that emit green fluorescence. After washed two times with incubation buffer, the green and red fluorescence were examined by flow cytometry (MilliporeSigma Co., Ltd., Burlington, MA, USA). The examination wavelength was 488 nm, and the emission wavelength was 530 nm.
Reverse-transcription and quantitative real-time polymerase chain reactions (RT-qPCR)
GCs were rapidly thawed and total RNA was isolated using the RNAprep Pure Micro Kit (Tiangen Biotech Co., Ltd., Beijing, China). The quality of RNA was checked at an absorbance of 260 nm/280 nm by Nanodrop-2000 (ThermoFisher Scientific, Waltham, MA, USA). Total RNA was reverse transcribed to cDNA using the PrimeScript™ RT reagent kit with gDNA Eraser (TaKaRa, Tokyo, Japan). Polymerase chain reaction (PCR) was performed using TB Green™ Premix Ex Taq™ II (TaKaRa) on a CFX96 real-time PCR detection system (Bio-Rad, Hercules, CA, USA) as follows: 95°C for 30 s, followed by 40 cycles of 95°C for 10 s, 60°C for 30 s, and 65°C for 5 s. The PCR system (20 µL) comprised RNase free dH2O (6.4 µL), cDNA (2 µL), forward primer (0.8 µL), reverse primer (0.8 µL) and 2 ×TB Green Premix Ex Taq II (10 µL). All PCR reactions were conducted in triplicate. Each experiment was repeated at least three times. Glyceraldehyde-phosphate dehydrogenase (GAPDH) was used as the internal control as indicated and fold changes were calculated by the 2-ΔΔCt method. Primers were designed and synthesized at Beijing Tsingke Biologocal technology (Beijing, China). Primers used in the RT-qPCR are shown in Table 1.
Table 1 Sequences of primers used in the qRT-PCR
Gene
|
Primer (5'→3')
|
Product size (bp)
|
Annealing temperature (℃)
|
FOXO1
|
F: TTTGCCCCAGATGCCTATAC
R: GGAGAGTCAGAAGTCAGCAAC
|
114
|
57.5
|
Bax
|
F: TTTCCGAGTGGCAGCTG
R: CAAAGTAGAAAAGGGCGACAAC
|
74
|
55.8
|
Bcl-2
|
F: GGATGCCTTTGTGGAACTGT
R: CACTTGTGGCTCAGATAGGC
|
135
|
57.4
|
caspase-9
|
F: TAACAGGCAAGCAGCAAAGT
R: ACCAAATCCTCCAGAACCAA
|
139
|
53.4
|
caspase-3
|
F: AGAACTGGACTGTGGCATTG
R: TAACCAGGTGCTGTGGAGTA
|
111
|
55.4
|
GAPDH
|
F: ACGGATTTGGTCGTATTGGG
R: CGCTCCTGGAAGATGGTGAT
|
214
|
57.4
|
Western blotting
Total protein was isolated from the GC samples using the RIPA lysis buffer (KeyGen Biotech. Co., Ltd.) containing Halt™ Protease Inhibitor Cocktail (Invitrogen, Karlsruhe, Germany) according to the manufacturer’s instructions. Total protein concentration was determined using a quantitative BCA protein kit (Thermo Scientific). The total proteins (60 µg/lane) were subsequently subject to 10% sodium dodecyl sulfate–polyacrylamide gel electrophoresis and transferred onto polyvinylidene fluoride membranes (EMD Millipore, Billerica, MA, USA). The membranes were then blocked with Tris-buffered saline with Tween-20® that contained 5% bovine serum albumin (Bio-Rad) for 1 h at room temperature and subsequently incubated with a primary antibody according to the manufacturer’s instructions at 4°C overnight. Specific primary antibodies included PI3K (1:2000; ab140307, Abcam, Cambridge, MA, USA), p-PI3K (Tyr607, 1:1000; ab182651, Abcam), Akt (1:10000; ab179463, Abcam), p-Akt (Ser473, 1:2000; ab81283, Abcam), FOXO1 (1:1000; 2880, Cell Signaling, Beverly, MA, USA), p-FOXO1 (Ser 256, 1:1000; 9461, Cell Signaling), Bax (1:1000; 5023, Cell Signaling), Bcl-2 (1:500; 01556, Wanleibio, Shenyang, China), caspase 9 (1:1000; 9502, Cell Signaling), cleaved caspase-9 (Asp330, 1:1000; 7237, Cell Signaling), caspase 3 (1:1000; ab32351, Abcam), cleaved caspase-3 (Asp175, 1:1000; 9661, Cell Signaling), and GAPDH (1:2000; 2188R, Bioss, Beijing, China). On the day after washing, the membranes were incubated with secondary antibodies for 2 hours at room temperature. SuperSignal® West Pico Trial Kit (ThermoFisher Scientific) was used for signal detection and the protein bands were visualized using a GelDoc XR densitometer (Bio-Rad). The relative intensities of each protein band were determined using the GAPDH band as an internal reference.
Concentrations of active caspase-9 and caspase-3 in GCs were measured by enzyme-linked immunosorbent assay (ELISA)
The cleaved caspase-9 and caspase-3 have bioactivity to induce apoptosis. The concentrations of active caspase-9 and caspase-3 in GC lysates were determined using human caspase-9 ELISA kit and caspase-3 ELISA kit (Elabscience Biotechnology Co., Ltd., Hubei, China), respectively, and a 450-nm Perlong DNM-9602G microplate spectrophotometer (Beijing Perlong New Technology Co., Ltd., Beijing, China) according to the manufacturer’s instructions. The amount of protein loaded in each well was the same (100 µg/well) and each sample was detected in duplicate. The intra- and inter-assay coefficients of variation for these values were < 5% and < 10%, respectively. The sensitivities of the caspase-9 and caspase-3 assays were 0.99 ng/mL and 0.19 ng/mL, respectively.
Statistical analysis
All data were statistically analyzed using SPSS 17.0 software (SPSS Inc., Chicago IL, USA). Continuous variables are expressed as means ± standard deviation. The normality of data distribution was assessed using Kolmogorov–Smirnov tests. Between-group comparisons were assessed using one-way ANOVA with post-hoc Bonferroni tests. Categorical data were compared using Chi-squared tests. Two-tailed P values < 0.05 were considered statistically significant.