Cell, cell culture and establishment of pulmonary metastasis cell lines
The mouse melanoma cell line B16F10 and the human melanoma cell line A375 were procured from Cell Resource Center of the Shanghai Institute for Biological Sciences (Chinese Academy of Sciences, Shanghai, China). Established post-metastasized melanoma cells, B16F10M and A375M were derived from the pulmonary metastases formed in mice by tail vein injection of pro-metastasized parental B16F10 and A375 cells, respectively, via a primary cell line establishment method[13,14]. This experimental metastasis refers to the injection of tumor cells directly to the systemic circulation and circumvents the invasion and extravasion.
The cells mentioned above were cultured in RPMI 1640 medium (Hyclone, Logan, UT, USA). Human pulmonary microvascularendothelial cells (HPMECs) were purchased from Cell Resource Center of the Shanghai Institute for Biological Sciences and cultured in ECM medium (Hyclone). All culture media was supplemented with 10 % (v/v) fetal bovine serum (FBS, Gemini Bioproducts, West Sacramento, CA) and 100 μg/mL penicillin/streptomycin (Sigma-Aldrich, St Louis, USA). Cells were maintained in a humidified atmosphere with 5 % CO2 at 37°C. Low oxygen concentration (1 % O2) was controlled by 245 whitley H35 HEPA hypoxystation (UK) or treated with YC-1 (MedChem Express, Princeton, NJ, USA). The cell lines were authenticated by Genetic Testing Biotechnology Corporation (Suzhou, China) using short tandem repeat (STR) markers. Cells were collected with 0.25 % trypsin (Hyclone) and counted by the ADAM MC Auto Cell Counter (NanoEnTek, Korea) before used.
Antibodies and reagents
The following antibodies were used: anti-NOL7 (#DF9720), anti-p-Smad2/3 (AF3367) from Affinity Biosciences (Cincinnati, OH, USA); anti-PCNA (#PC10), anti-E-cadherin (#3195), anti-N-cadherin (#13116), anti-Vimentin (#5741), anti-β-catenin (#8480), anti-p21 (#2947), anti-p27 (#3686), anti-CDK2 (#2546), and anti-MMP9 (#13667), anti-Smad2/3 (D7G7) from Cell Signaling Technologies (Beverly, MA, USA); anti-AKT 1/2/3 (#sc8312), anti-p-AKT 1/2/3 (Ser 473, #sc33437), anti-ERK 1/2 (#sc135900), anti-p-ERK 1/2 (Thr 202/Tyr 204, #sc16982) and anti-Twist (#sc81417) from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA); anti-JNK (#WL01295), anti-p-JNK (#WL01295), anti-HIF-1α (#WL01607), anti-Gsk3β (#WL0146), anti-caspase 3 (#WL02117), anti-caspase 9 (#WL01551), anti-bad (#WL02304), anti-bax (#WL01637), anti-survivin (#WL03492), anti-cyclin A (#WL01841) and anti-cyclin E (#WL01072) from Wanlei Biotechnology (Shenyang, China); anti-Fibronectin (#40932) from SAB Technology (Danvers, USA); anti-TSP-1(A2125), anti-Rb (A3618) from ABclonal Technology (Wuhan, China); β-actin (AC001-R), secondary antibody goat anti-mouse-IgG horseradish peroxidase and goat anti-rabbit-IgG horseradish peroxidase from Dingguochangsheng Biotechnology (Beijing, China).
NOL7 siRNA (sense, 5’-3’, GCUGUAUUAGAGCAGCUAATT) for B16F10 cells, NOL7 siRNA (sense, 5’-3’, GGAAAUGACUCCAAGAAAGATT) for A375 cells, corresponding negative control siRNA (UUCUCCGAACGUGUCACGUTT) and NOL7 guide RNA sequences (sgRNA, sense, 5’-3’, CACCGACCGCGAGCGTCTCGCGCCC) for A375 cells were synthesized by Sangon Biotech (Shanghai, China).
Gene constructs and generation of stable transfectants
Specific siRNAs were used for the transient knockdown of NOL7 in B16F10 and A375 cells. CRISPR/Cas9 system was operated to generate the stable NOL7-knockout A375 cells based on a previous protocol[15]. In short, sgRNA for A375 cells was designed by a CRISPR design platform (Zhang Feng Lab, http://crispr.mit.edu/) and then cloned into the PX458 vector. The empty vector was used as a control. These oligo duplexes were transfected into cells via Lipofectamine 3000 Kit (Invitrogen, MA, USA) through standard procedure. Furthermore, stable NOL7-knockout cells were sorted through GFP-activated cell sorting method by flow cytometer (FACS Aria III, BD, San Diego, USA).
Cell proliferation assay
For cell growth kinetic assay, NOL7-knockdown melanoma cells or control cells were plated in 96-well plates (5×103 cells/well). Cell proliferation was monitored every day for 5 days using cell counting kit-8 assay (Promega, Sunnyvale, CA) according to the manufacturer’s protocol.
For cell spherical growth assay, A375-sgNOL7 cells or control cells were put into 96-well microplates with ultralow attachment surfaces (Corning). Visually 3D cell spheroids were formed after 24 h, and their growth was observed and photographed at regular time intervals by phase contrast microscopy to monitor the tumor spheres diameter.
Colony formation assay
A total of 1000 target cells per well were plated in 6-well plates. After culture in complete medium for 14 days, cell colonies were washed three times with PBS and fixed in 4 % paraformaldehyde for 20 min and then, stained with a 0.2 % crystal violet (Sigma-Aldrich) solution for 40 min at room temperature. An overall image of the cell colonies was mapped by Canon scanner (Shanghai, China). Colonies with 50 or more number of cells were counted under a light microscope (Zeiss, Oberkochen, Germany).
Cell cycle assay
Suspended cells were washed twice with precooled PBS and fixed in 70 % ethanol for 24 h at 4°C. The cells were then stained with a fluorescent solution (1 % (v/v) Triton X-100, 0.01 % RNase, 0.05 % PI (Sigma-Aldrich)) for 30 min at 37°C in the dark. Ten thousand events were acquired by flow cytometry to measure the cell cycle distribution. Data were processed by Modfit software 3.2.
Cell apoptosis assay
Using an Annexin V-FITC/PI kit (KeyGen Biotech, China), cell apoptosis was detected by flow cytometry. Target cells at a density of 3×105 cells per well were plated in 6-well plates in triplicate. The cells were preprocessed under the following conditions: one group of cells was cultured in complete medium; one group of cells was treated with 10 μM paclitaxel (Meilunbio, Dalian, China) and another group of cells was kept in a suspended state or in FBS-free medium. All of these cells were collected separately after 24 h, and stained with Annexin V-FITC/PI for 15 min under the lucifuge condition. At least 104 cells from each specimen were examined by flow cytometry and data were analyzed by FlowJo software 10.0.
Domestication of anoikis-resistant A375 cells
Anoikis-resistant A375 cells were generated through continuous cycle of culturing on normal (adherent) and soft agar (Sigma-Aldrich) -coated cell culture well (suspended). Briefly, A375 cells were plated in soft agar-coated wells and maintained under suspended conditions for the indicated time intervals. After 24 h incubation, cells were washed and transferred to normal cell culture wells. We measured the anoikis-resistance of A375 cells by analyzing the cells apoptosis.
In vitro cytotoxicity assay
A MTT (3-(4,5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide, Sigma-Aldrich) assay was used to assess cell sensitivity to paclitaxel, an broad spectrum anticancer drug[16]. The cells were plated in 96-well plates at a concentration of 104 cells/0.1 mL per well. The cells were treated with various concentrations (0-100 μM) of paclitaxel for 24 h. Finally, the culture medium was replaced with MTT solution (5 mg/mL in phenol red or serum-free medium) and incubated for an additional 4 h. Formazan crystals formed by metabolically viable cells were dissolved in 150 μL DMSO. Absorbance at 490 nm was recorded by an Infinite M200 Pro microplate reader (Tecan, Hombrechtikon, Switzerland).
Motility assay
Cell motility was measured by wound healing assay. Monolayer cells in 6-well plates were scratched with a 100 μL pipette tip (t = 0 h) and maintained in 1640 medium containing 1 % FBS. The wounded area was photographed at 0 h and 24 h by a light microscope (Zeiss). Cell mobility was quantified as the difference value between the width of the wound at a given time and the initial width of the wound.
Migration and invasion assays
Falcon cell culture inserts with 8 μm pores (Corning, Cambridge, MA, USA) were placed in 24-well plates. To measure invasion, the upper chamber of a Transwell apparatus was precoated with Matrigel (Corning). Cells in 200 μL culture medium containing 1 % BSA were plated into the upper chamber and 800 μL culture medium containing 20 % FBS was added to the lower chamber. After incubation for 24 h, the noninvasive cells that adhered to the upper chamber were wiped away, while the invasive cells adhered to the lower chamber were fixed with 4 % paraformaldehyde for 30 min and stained with crystal violet for 40 min. The invasive cells per field were assessed by a fluorescence microscope (Zeiss). To measure cell migration, the upper chamber of the inserts was not precoated with Matrigel.
Adhesion assay
To measure cells adhesion to endothelial cells, monolayer HPMECs were precultured in 6-well plates and stimulated with IL-1β (10 ng/mL, Sigma-Aldrich) for 4 h. Subsequently, the target cells labeled with rhodamine 123 (Sigma-Aldrich) came into contact with the surface of HPMECs and oscillated at 200 rpm for 30 min. After that, the cells that didn’t attach to the HPMECs were washed away gently. The cells adhered to HPMECs were photographed and counted by phase-contrast light microscopy (Zeiss). Similarly, to measure adhesion to the ECM, rhodamine 123-labeled target cells were plated into 6-well plates with fibronectin-coated (Sigma-Aldrich) and incubated without shaking for 30 min.
Western blotting
Cell lysates were extracted by RIPA lysis buffer containing 1 μM PMSF at 4℃. Then each equal quality of each sample was separated by SDS-PAGE and transferred to PVDF (Bio-Rad, USA) membranes. After blocking in 5 % skim milk solution for 2 h at room temperature, the PVDF membranes were incubated with dilute primary antibodies overnight at 4°C and secondary antibodies at room temperature for 2 h. Protein blots were visualized using the hypersensitive chemiluminescence kit (Wanlei, Shenyang, China) through ChemiDoc XRS system (Bio-Rad). Protein expression levels were analyzed by Image Lab analysis software 5.0 (Bio-Rad). β-actin was served as the internal control.
Quantitative Real-Time PCR
Total RNA was extracted using TRIzol reagent (Invitrogen), and 100 ng of each sample was then used as template for reverse transcription to cDNA via a PrimeScript® reverse transcription kit (Takara, Japan) in accordance with the manufacturer’s guide. PCR was performed by SYBR® Premix Ex Taq™ PCR kit (Takara) on a CFX96™ real-time PCR detection system (Bio-Rad). The threshold cycle values were used to quantify the relative mRNA expression and normalized to β-actin. Primer sequences used are listed in Supplementary Table 1.
Mice and in vivo mouse study
The mice experiment was guided and approved by the Institutional Animal Care and Use Committee (IACUC) of Fuzhou University, the approval number is SYXK-2019-0007. Female BALB/c nude mice 6-8 weeks old and female C57BL/6 mice 6-8 weeks old were obtained from Slac Animal Inc (Shanghai, China). BALB/c nude mice were housed in clean and sterile cage (Sujingantai Biotechnology, Suzhou, China) and fed with sterilized rat food (Fuzhou Wushi Animal Center, Fuzhou, China). and sterilized water. C57BL/6 mice were housed in clean cage and fed with usual rat food (Fuzhou Wushi Animal Center) and clean water. All mice were accessed to food and water ad libitum. The animal house was kept in a 12 h-day/night cycle with lights on at 7:00 a.m and in a temperature (26 ± 1°C) and humidity (50 ± 10 %). All mice were randomised into each group by picking random numbers. The mice in all experiments involved in this manuscript were not given any drugs or anesthetic. We have tired our best to minimize animals’ pain and suffering.
To obtain the pulmonary metastases of melanoma described above, B16F10 cells (5×104 cells in 0.1 mL PBS per mouse, 5 mice per group) were injected into the tail vein of C57BL/6 mice and A375 cells (3×106 cells in 0.1 mL PBS per mouse, 5 mice per group) were injected into the tail vein of BALB/c nude mice, respectively. After 7 weeks, all mice were killed by cervical dislocation in a biological safety cabinet (Sujingantai Biotechnology) and the pulmonary metastases were collected for further analysis.
To assess cell metastatic potential, A375-sgNOL7 cells (3×106 cells in 0.1 mL PBS per mouse, 5 mice per group) were injected into tail veins of BALB/C nude mice. Mice injected with A375-control cells (3×106 cells in 0.1 mL PBS per mouse, 5 mice per group) were used as control group. After 7 weeks, all mice were sacrificed by cervical dislocation and dissected in a biological safety cabinet. The pulmonary metastatic nodules were counted for further analysis.
To assess the cell tumorigenicity, A375-sgNOL7 cells (6×106 cells in 0.1 mL PBS per mouse, 5 mice per group) were subcutaneously injected into the left axillae of BALB/C nude mice. Mice injected with A375-control cells (6×106 cells in 0.1 mL per mouse, 5 mice per group) were used as control group. About 7-10 days later, the subcutaneous tumor grew and tumor volumes were measured every alternate day. After 4 weeks, all mice were killed by cervical dislocation and dissected in a biological safety cabinet. The tumors were collected for further analysis.
Pathological analysis
Excised neoplasms and lung tissues were embedded in paraffin and sliced into sections at thickness of 5 μm. Afterward, histological slices were subjected to hematoxylin and eosin (H&E) staining and imaged by a light microscope (Zeiss).
Immunohistochemistry (IHC)
For validation of the clinical significance of NOL7 in melanoma. Various clinical samples from melanoma patients (normal skin, n = 5; nevus, n = 5; primary melanoma, n = 32; metastases, n = 10) were purchased from Shanghai Zhuoli Biotechnology Co., Ltd (Shanghai, China). NOL7 expression in these samples was evaluated by IHC analysis with the assistance of Shanghai Zhuoli Biotechnology Co., Ltd. The immunostaining score of NOL7 in tumor tissues was quantified by the H-score (histochemical score) based on the staining intensity and heterogeneity under a Nano Zoomer S210 (Hamamatsu, Japan). The percentage of immunostaining and the staining intensity were scored as 0, negative; 1, weak; 2, moderate; and 3, strong. H-score was calculated using the following formula:
H score = (percentage of cells of weak intensity×1) + (percentage of cells of moderate intensity×2) + (percentage of cells of strong intensity×3)
For analysis of NOL7 expression in melanoma formed in mice. NOL7 expression was evaluated by IHC analysis with the assistance of Wuhan Servicebio Co., Ltd. In brief, paraffin embedded sections were fixed in formaldehyde, and a heat-mediated antigen retrieval step in citrate buffer was performed. The samples were blocked in BSA solution for 2 h at 37°C and incubated with anti-NOL7 antibody overnight at 4°C. An horserdish peroxidase conjugated goat anti-rabbit antibody was used as the secondary antibody. NOL7 protein was visualized by AEC staining and examined under the confocal fluorescence microscope (Nikon Eclipse C1, Nikon).
Statistical analysis
Date for all experiments was managed using GraphPad Prism software 5.0 and represented as the mean ± s.d. A paired t test was used for two group comparison, and a one-way ANOVA was used for multiple group comparison. P < 0.05 was considered representative of a significant difference and statistical significance was symbolized by asterisks corresponding to * P < 0.05, ** P < 0.01 and *** P < 0.001.