Cell culture and generation of Sf3b1 mutant R625H cells
The GH3 rat pituitary cell line was purchased from the American Type Culture Collection (CLL-82.1; Manassas, VA, USA) and was cultured in Ham’s F12K medium with 2.5% fetal bovine serum (FBS) and 15% horse bovine serum (Gibco, Waltham, MA, USA). MCF7 and HEK293T cells were obtained from the National Infrastructure of Cell Line Resource (Beijing, China) and were cultured in DMEM (Gibco) supplemented with 10% FBS. PRL levels in cell culture supernatant were detected using an ELISA kit (BioVision, Milpitas, CA, USA) according to the manufacturer’s instructions.
The CRISPR/Cas9 gene editing system was used to generate the Sf3b1 p.R625H (c.G1874A, c.A1875T) mutant GH3 cell line. A Sanger Centre CRISPR webtool (http://www.sanger.ac.uk/htgt/wge/) was used to identify two small guide RNAs (sgRNAs). Cas9/sgRNA-mediated DNA double-strand break and homologous recombination contributed to generated the specific mutation. The 5′ sgRNA sequence was 5′-GGCAAGATTCCTTCCTCA-3′ and the 3′ sgRNA sequence was 5′-GGTTAAGAGTACTGTTGTC-3′. The sgRNA pairs were cloned into a wild-type spCas9 and sgRNA expression plasmid. The donor plasmid (CL-GJ-016-puro-ΔTK) consisted of a puromycin resistance cassette flanked by two loxP sites and ~ 1 kb homology arms at both ends of the c. 1874 G > A, c. 1875 A > T mutant exon 14 (Wuhan Genecreate Biological Engineering Co. Ltd., Wuhan, China) (Fig. 1). The plasmid was constructed according to previously published protocols and confirmed by sequencing. The Neon Transfection System (Thermo Fisher Scientific, Waltham, MA, USA) with pulse voltage of 1000 V for 40 ms was used to co-transfect GH3 cells on a 10-cm plate with 1 µg of CL-GJ-016-puro-TK and 3 µg of Cas9/sgRNA. The cells were screened with puromycin (0.5 µg/mL) starting 48-h post-transfection. Thereafter, single-cell cloning was performed in a 96-well plate. Sanger sequencing was performed using genomic DNA to confirm the specific mutations (DIA-UP Biotech, Beijing, China). Primers were designed to amplify exon 14 of the Sf3b1 gene (Table S1).
Adenoviral constructs and primary cell culture of human prolactinomas
The adenoviral constructs for mutant SF3B1R625H and wild-type SFB31 were generated by BAC Biological Technology (Beijing, China), and primary cultures of human prolactinoma cells were prepared as described in our previous publication[16]. Tumor cells were infected with adenovirus at a multiplicity of infection of 100, and then harvested for quantitative (q)PCR 48-h later.
RNA sequencing
For GH3 cells, total RNA was extracted using the AllPrep® DNA/RNA Mini kit (QIAGEN, Hilden, Germany) according to the manufacturer’s instructions. Sequencing libraries were generated using NEBNext® Ultra™ Directional RNA Library Prep Kit for Illumina® (NEB, Ipswich, MA, USA). The libraries were sequenced on an Illumina platform, and 150-bp paired-end reads were generated. Clean reads were mapped to the Rat Rnor_6.0 genome using Hisat2 (v2.0.5) to obtain read counts, fragments per kilobase per million mapped fragments (FPKM), and transcripts per million (TPM) for each sequenced gene.
Differential alternative splicing and differential gene expression analyses
Differentially expressed genes (DEGs) were identified using DESeq2 using P < 0.05 and absolute value of fold change ≥ 1.5. For alterative splicing analysis, rMATS (version 4.0.2)[17] was used to identify alternative splicing events by quantifying exon-exon junction spanning reads on annotated splice junctions present in the rat GENCODE Rnor_6.0 assembly. Differentially spliced mRNAs were defined as FDR < 0.05 and a minimum inclusion level difference > 10% or < − 10%. Three mutant GH3 replicates and three wild-type replicates were compared. Data of two prolactinoma patients with the SF3B1R625H mutation and two wild-type cases were selected for alternative splicing analysis, and two mutant and 13 wild-type cases from our previously reports were selected for differential expression analysis[16]. Basic clinic data of these patients are listed in Table S2. Functional enrichment analysis was performed using Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) to predict the biological functions of DEGs and differentially spliced transcripts. The GO terms analyzed included biological process, cellular components, and molecular functions. A KEGG pathway with a P value < 0.05 was considered to be significant.
Whole-genome sequencing and variant identification
Genomic DNA was extracted from wild-type and mutant GH3 cells using the Blood & Cell Culture DNA Mini Kit (QIAGEN). DNA was sequenced using an Illumina HiSeq 2000, generating 150-bp paired reads. Reads were aligned to the Rat Rnor_6.0 genome using Burrows–Wheeler Aligner (BWA)[18]. SAMtools was used to generate BAM files[19]. Visual inspection of the Sf3b1 R625H mutation was performed using the Integrative Genomics Viewer[20].
Minigene assay
A DNA fragment containing the DLG1 exon 22 genomic sequence with 151 bp of flanking intron 21 and 271 bp of flanking intron 22 was inserted between the KpnI and EcoRI restriction sites of the pcMINI vector to produce the DLG1 minigene construct. Sanger sequencing was performed to confirm the sequence of the inserted fragment. Briefly, the DLG1 minigene and an adenoviral vector were co-transfected into 293T cells. After 48 h, RNA was harvested for PCR analysis. PCR products were separated by 2% agarose gels and confirmed by Sanger sequencing. The primers used for the spliced products were: 5′-CTAGAGAACCCACTGCTTAC-3′ (forward) and 5′-TAGAAGGCACAGTCGAGG-3′ (reverse).
Transfection
RIBOBIO (Guangdong, China) synthesized small interfering (si)RNA duplexes; siRNA sequences of rat and human DLG1 are listed in Table S2. The Dlg1 overexpressed plasmid, pLV-hef1a-Puro-WPRE-CMV-Dlg1-3×FLAG, was constructed by Beijing Syngentech Co., Ltd. (Beijing, China). All transfections (siRNAs and overexpression plasmids) were performed using Lipofectamine® 3000 Transfection Reagent (Invitrogen, Carlsbad, CA, USA) according to the manufacturer’s protocols. Cells were transfected with siRNA or plasmids for 48–72 h. and then collected for qPCR and western blot assays.
Reverse transcription PCR and qPCR
The RNeasy Mini Kit (QIAGEN) was used to extract total RNA and the High Capacity cDNA Reverse Transcription Kit (Thermo Fisher Scientific) was used to generate cDNA, both according to the manufacturers’ instructions. For reverse transcription PCR, amplification was performed with I-5 High-Fidelity Master Mix (MCLAB, San Francisco, CA, USA) and specific primers. PCR products were separated on 1–3% agarose gels. All qPCR assays were performed using Power SYBR™ Green PCR Master Mix (Thermo Fisher Scientific) and were analyzed using QuantStudio 3 and 5 systems (Applied Biosystems, Waltham, MA, USA). The comparative Ct method was used to evaluate relative gene expression. The primers used in this study are listed in Table S3.
Scanning electron microscopy (SEM)
SEM was used to examine the external morphological alterations of Sf3b1 wild-type and mutant GH3 cells. Cells were collected and fixed with 2.5% glutaraldehyde (Solarbio, Beijing, China) at 4°C overnight for SEM preparation. After rinsing with PBS and sterile water, the cells were dehydrated using an ethanol gradient. The samples were coated with gold after critical point drying, and electron micrographs were taken on a Hitachi SU8020 SEM (Tokyo, Japan).
Cell migration and wound-healing assays
Cell migration culture dish inserts from Ibidi (Martinsried, Germany) were used to conduct wound-healing assays. After 24 h of transfection, cells were seeded into the chambers of the culture dish inserts. The inserts were removed on the next day, and fresh culture medium was added to each well. Scratches were photographed at different points in time using a Zeiss microscope (Oberkochen, Germany).
Sf3b1 mutant and wild-type GH3 cells were seeded onto Imagelock 96-well plates (Essen Bioscience, Ann Arbor, MI, USA). The IncuCyte® Wound Maker (Essen Bioscience) was used to make uniform wounds in a monolayer of confluent cells. Phase contrast imaging was performed every 12 h for a total of 96 h. Images were analyzed using IncuCyte® S3 2018B-2019A software (Essen Bioscience), and data were analyzed using GraphPad Prism7 (GraphPad Software, Inc., La Jolla, CA, USA).
Transwell assays
Transwell plates and Matrigel-coated transwell plates (Corning-Costar, Corning, NY, USA) were used to determine cell migration and invasion capabilities, respectively. Briefly, HEK293T cells resuspended in serum-free medium were inoculate into the upper chamber, and DMEM with 10% FBS was placed in the bottom chamber. Following 24 h of culture, cells on the bottom surface of the chamber were stained with crystal violet and counted under a microscope (Zeiss).
Western blot analysis
Protein samples were separated on 8–10% Bis-Tris SDS-PAGE gels and transferred to polyvinylidene fluoride membranes (Merk, Kenilworth, NJ, USA). All primary antibodies (Table S4) were diluted in TBST containing 1% bovine serum albumin (BSA) and were incubated with the membranes overnight at 4°C. Immunoreactive bands were visualized by chemiluminescence.
Immunohistochemistry
Human prolactinoma tumor specimens were used to examine DLG1, E-cadherin, and Snail protein levels in SF3B1 wild-type and mutant tumor tissues. The primary antibodies used are summarized in Table S4. Immunohistochemistry was performed by the Leica Bond Polymer Refine Detection system (Leica Biosystems, Wetzlar, Germany). All slides were scanned into digital images, and expression was examined using Aperio AT2 (Leica Biosystems). Staining intensity was scored as 0 (negative), 1 (weak), 2 (moderate), or 3 (strong). The percentage of immunostaining was recorded, and H-scores were calculated using the formula: H-score = 1 × (% weakly stained cells) + 2 × (% moderately stained cells) + 3 × (% strongly stained cells)], ranging from 0 to 300.
Rat prolactinoma model
Rat pituitary tumors were induced by subcutaneously implanting 1-cm silastic capsules containing 10 mg of 17-β estradiol in 4-week-old female F344 rats. Prolactinomas were induced by 17β-estradiol for 5 weeks, as described in our previous report[21]. All experimental protocols were approved by the Animal Use and Care Committee of Beijing Tiantan Hospital. Five weeks later, the prolactinomas were validated via 7.0-T magnetic resonance imaging (MRI) before intra-pituitary injection. The rats were anaesthetized and then 1 µl of adenovirus vector control, wild-type SFB31, or SF3B1R625H was stereotactically injected into each bilateral tumor. A second MRI examination was performed 2 weeks later, and all rats were sacrificed to collect tumor tissues for further analysis.
Immunofluorescence staining of rat tumor tissues
First, rats were anaesthetized and heart-perfused with 4% formalin. Tumor tissues were then collected and immersed in 10% sucrose for 2 h, and then incubated in 30% sucrose overnight before freezing in OCT compound. Prior to immunofluorescence staining, frozen 5-µm-thick sections were fixed in ice-cold acetone. Tissue slides were blocked with goat serum and incubated with primary antibodies overnight at 4°C (summarized in Table S4). After washing with PBS, the slides were incubated in Alexa Fluor 488 and 594 secondary antibodies (Invitrogen) for 1 h at room temperature; DAPI was used to visualize nuclei.
Phalloidin staining and confocal microscopy
Cells were plated on confocal dishes coated with poly-L-lysine. After incubation at 37°C for 24 hours, the cells were fixed with 4% paraformaldehyde for 30 min, and then stained with 5 µg/mL Alexa Fluor 488-phalloidin (Invitrogen) to reveal filamentous actin (F-actin) in PBS for 40 min at 37°C. DAPI was used to visualize nuclei, and images were captured by confocal laser-scanning microscopy (Zeiss).
Statistical analysis
All statistical analyses were performed using R v3.4.1 (https://www.r-project.org/) and Prism 7 (GraphPad Software, Inc.). All experiments were performed with at least three biological replicates, and all quantitative data represent mean ± standard deviation (SD). Statistical significance was determined by unpaired Student’s t test (two groups) or one-way ANOVA (multiple groups). P < 0.05 was considered statistically significant.