Patient samples and cell lines
All the samples were obtained following transsphenoidal surgery performed at Beijing Tiantan Hospital from June 2018 to June 2019. Fresh tumor samples were stored in liquid nitrogen. 45 corticotroph adenomas, 79 gonadotroph adenomas, 17 lactotroph adenomas and 31 somatotroph adenomas from the study population (age range, 20–75 years) were diagnosed according to the 2017 World Health Organization classification of tumors of endocrine organs. The study protocols were approved by the Internal Review Board of Beijing Tiantan Hospital, which was affiliated with Capital Medical University, and conformed to the ethical guidelines of the Declaration of Helsinki (No. KY2016-035-01).
The GH3 and MMQ cell lines (ATCC, Manassas, VA, USA) cultured in a humidified incubator at 37°C and 5% CO2 in F-12K medium (ATCC) supplemented with 2.5% fetal bovine serum and 10% horse serum. The ATT20 cell line (ATCC) was cultured in a humidified incubator at 37°C and 5% CO2 in DMEM (ATCC) supplemented with 10% fetal bovine serum.
RNA extractions, sequencing, transcriptomic data processing and analysis
For the RNA extractions, patient samples were processed with an AllPrep® DNA/RNA Mini kit (Qiagen, Hilden, Germany) according to the manufacturer’s instructions. The quantity and quality of the RNA was evaluated by an RNA Nano6000 assay kit (Aligent Technologies, CA, USA) (RIN>6.8). RNA (3 μg/sample) was used for the RNA preparations, and the ribosomal RNA was removed using an Epicentre Ribo-zero™ rRNA Removal Kit (Epicentre, Madison, WI, USA). The sequencing library was generated using the NEBNext® Ultra™ Directional RNA Library Prep Kit (NEB, Ispawich, USA). The library fragments (150~200 bp) were purified by the AMPure XP system (Beckman Coulter, Beverly, USA) and then assessed by the Agilent Bioanalyzer 2100 system. The libraries were sequenced on an Illumina HiSeq X platform, and then, 150-bp paired-end reads were generated. Reads containing adapters, reads containing poly-N and low-quality reads were removed. The paired-end clean reads were aligned to the human reference genome (hg19) using Hisat2 (v2.0.5) [21]. HTSeq (v0.11.2) was used to count the read numbers mapped to each gene [22]. The R package limma was used to analyze the quantitative differentiation between two identified groups [23]. KEGG pathway enrichment and GO term results were exported from the Metascape website with filtered differential gene data input [24]. The R package ClusterProfiler was used to process the GSEA analysis [25].
Tissue microarray construction and immunochemistry staining
The formalin-fixed, paraffin-embedded tissue blocks were sectioned. Three core biopsies (2.0 mm in diameter) were selected from the paraffin-embedded tissue. The cores were transferred to tissue microarrays using a semiautomated system (Aphelys MiniCore, Mitogen, UK). The microarrays were cut into 4-μm sections and incubated with anti-DLK1 (rabbit monoclonal, 1:600, ab210471, Abcam), anti-PIT1(mouse monoclonal, 1:500, sc393943, Santa Cruz) and anti-SSTR2 (rabbit monoclonal, 1:400, ab134152, Abcam) primary antibodies. BondTM Ploymer Refine Detection (Leica Biosystems, DS9800) was used for the detection of the primary antibodies. The slides were scanned into digital pictures, and expression was examined using Aperio AT2 (Leica Biosystems). The H-score was obtained by multiplying the staining intensity by a constant to adjust the mean to the strongest intensity [H-score = 3×(percentage of strong staining)] (1.0%, weak; 2.0%, moderate; 3.0%, strong) to yield a score ranging from 0 to 300.
Cell proliferation and colony formation assay
The number of cells in suspension was adjusted to 1×105/ml, and 100 µl cell suspension was plated into each well of a 96-well plate. After overnight incubation, anti-DLK1 monoclonal antibody blockade (1 μg/ml, 5 μg/ml, and 20 μg/ml) or siMEG3 fragments (Supplementary Table 1) were added.
The cells were then cultured for 24 h, 48 h and 72 h; 20 µl 3-(4,5-diethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium, inner salt (MTS) solution was then added into each well and incubated for an additional 4 h. The absorbance at 490 nm was measured with an ELISA plate reader (Thermo, USA). For the colony formation assay, a total of 200 cells were plated into each well of a 24-well plate and maintained for 7 days. The colonies were then fixed with 4% formaldehyde for 30 min and stained with 0.2% crystal violet for 10 min. Triplicate wells were measured in each treatment group.
Immunofluorescence and confocal microscopy
For the immunofluorescence experiments, GH3 cells were seeded on poly-L-lysine-coated confocal dishes, and an anti-DLK1 antibody (5 μg/ml) was added. After 24 h in culture, the cells were fixed with 4% paraformaldehyde for 30 min, washed with PBS, and permeabilized with 0.2% Triton X-100 in PBS for 15 min at room temperature. The fixed cells were incubated with an anti-DLK1 (1:600) antibody and anti-PIT1 antibody (1:500) overnight at 4°C followed by incubation with the fluorescently (Alexa Fluor 488 and 594) labeled secondary antibody (1:300) for 1 h at room temperature. The cell nuclei were visualized by DAPI counterstaining (Invitrogen).
ELISA assay
The adrenocorticotropic hormone (ACTH), GH, IGF-1, and prolactin (PRL) levels in 10 μl cell culture supernatant were detected by ELISA kits (APPLYGEN) according to the protocol. The absorbance of each well at 450 nm was measured using an ELISA plate reader (Thermo Fisher).
Reverse transcription and quantitative PCR
Microarray hybridization and qRT-PCR were performed as previously described [26]. The total RNA of 30 samples was extracted and purified using the Rneasy®Mini Kit (QiaGen, Hilden, Germany) following the manufacturer’s instructions. RT-qPCR was performed on a QuantStudio5 (Applied Biosystems, Singapore) with primers of each genes (Supplementary Table 2). The fold-change in the differential expression of each gene was calculated using the comparative CT method (2−∆∆CT method) in the R package PCR, with GAPDH as the reference gene [27].
SDS-PAGE and western blot analyses
Samples (up to 10 mg) were lysed in lysis buffer containing 1% Nonidet P-40 (Calbiochem, Merck, Darmstadt, Germany) and protease and phosphatase inhibitor cocktails (Roche, IL, USA) overnight at 4°C. The total extracts were centrifuged at 12,000 g for 30 min at 4°C, and the protein concentration was determined by the BCA method (Pierce Biotechnology, IL, USA). A total of 20 μg of protein per lane was loaded onto 10% Bis-Tris SDS-PAGE gels, separated electrophoretically, and blotted onto polyvinylidene fluoride membranes (Merck). The blots were incubated with anti-ACTH (rabbit polyclonal, 1:2,000, AB74976), anti-GH (mouse monoclonal, 1:1,000, ab155276), anti-PRL (rabbit polyclonal, 1:1,000, ab64377), anti-DLK1 (1:2,000, Abcam), anti-PIT1(1:2,000, Santa-Cruz), anti-phospho-4EBP1 (rabbit polyclonal, 1:2,000, AF3432, Affbiotech), anti-4EBP1 antibody (mouse monoclonal, 1:1,000, 60246-1, Proteintech), anti-phospho-P70S6K (rabbit polyclonal, 1:1,000, AF3228, Affbiotech), anti-P70S6K antibody (rabbit polyclonal, 1:1,000, 144851-AP, Proteintech), anti-phospho-mTOR-2448 (rabbit monoclonal, 5536s, 1:1,000, CST) and anti-mTOR (rabbit monoclonal, 1:1,000, 2983s, CST) antibodies, followed by a secondary antibody (1:8,000) tagged with horseradish peroxidase (Santa Cruz Biotechnology). The blots were visualized by enhanced chemiluminescence, and densitometry was performed using a fluorescence image analyzer (Amersham Imager 600, GE, MA, USA). GAPDH was used as the loading control.
Statistical analysis
All the statistical analyses were conducted using SPSS Statistics Version 22 (IBM Corporation, Armonk, New York, USA). Unpaired Student’s test and chi-square (Fisher’s exact) test were used to compare quantitative and qualitative data. The P value of less than 0.05 was considered significant. The R package pROC was used to compute ROC curves, AUC, and P values to evaluate the predictive accuracy of selected genes [28].