Cell culture
The CT26 murine colorectal adenocarcinoma and B16-F10 murine melanoma cell lines were obtained from the American Type Culture Collection (Manassas, VA, USA). CT26 and B16-F10 cells were cultured in RPMI-1640 and DMEM medium (Invitrogen, Carlsbad, CA, USA), respectively, with 10% heat-inactivated fetal bovine serum (Hyclone, Logan, UT, USA) and 2 mM L-glutamine (Invitrogen). Stable clones of CT26 cells expressing luciferase (CT26-Luc) were generated by lentiviral transduction with the North American firefly luciferase gene under the control of the SV40 promoter. CT26-Luc cells were further passaged in the previously-mentioned medium containing G418 (400 µg/ml; Sigma-Aldrich, St. Louis, MO, USA).
Lung metastatic mouse model and treatments
Four-week-old male BALB/c mice or C57BL/6 mice were acquired from the Animal Resource Center of the National Science Council of Taiwan (Taipei, Taiwan). All animal experiments have been approved by the Animal Ethics Committee of China Medical University, Taiwan (CMUIACUC-2021-018). We intravenously administered CT26-Luc (2 ⋅ 106) into the tail veins of mice to induce lung metastasis and assessed lung metastatic tumor growth by the IVIS 200 imaging system (Xenogen Biosciences, Cranbury, NJ) at the indicated time points. Alternatively, B16-F10 (1 ⋅ 106) cells were intravenously injected into the tail veins of mice to induce pulmonary dissemination and scored the number of black lung melanoma nodules on the surface of each lobe using a dissecting microscope (Motic China Group Co., Ltd). Before irradiation, tumor-bearing mice were intraperitoneally anesthetized using a mixed solution of Zoletil™ (4 mg/kg) and Ropum™ (1 mg/kg), then single limb irradiation (SLI) treatment was performed by a linear accelerator (Clinac 1800, Varian Associates, Inc., Palo Alto, CA, USA) with 25x25 cm field size and an SSD of 100 cm for electron beams with energies of 6 MeV. Left hind limbs of tumor-bearing mice were exposed to 5 Gy with a bolus of 0.5 cm to improve dose homogeneity. For immune checkpoint inhibitor therapy, the anti-mouse PD-1 mAb (200 µg/mouse; clone RMP1-14, BioXcell), anti-mouse PD-L1 mAb (150 µg/mouse, clone 10F.9G2, BioXcell) were intraperitoneally administered on days 0, 2, and 4 post-SLI treatment. For EPO experiments, mouse EPO protein (10 µg/kg, #587602, BioLegend, San Diego, CA, USA) was retro-orbitally injected on days 0, 2, and 4 post-SLI treatment, and anti-EPO mAb (7.5 mg/kg, clone 148438, R&D Systems, Minneapolis, MN, USA) was intraperitoneally administered on days 0 post-SLI treatment. Mice were sacrificed on day 4, 8, and 17 for TME assessment.
Hemogram and biochemistry analyses
Murine blood samples were obtained by facial vein bleed and collected in tubes containing 20 IU/ml of heparin. Plasma was further separated by spinning at 1,000 x g at room temperature for 20 min. White blood cell (WBC) counts were quantified by an automatic Coulter counter (HEMAVET HV950; Drew Scientific, Inc., Dallas, TX). Functional indicators including alanine aminotransferase and creatinine were colorimetrically assessed using Fuji Dri-Chem Slide and analyzed by Fuji Dri-Chem 3500i Biochemistry Analyzer (Fujifilm Ltd, Japan) according to instructions of the manufacturer.
Flow cytometric analysis
Mice with lung metastasis were euthanized via intraperitoneal injection of a mixed solution of Zoletil™ (4 mg/kg) and Ropum™ (1 mg/kg). Whole lungs were removed and digested with collagenase A (1.5 mg/ml) and DNase I (0.4 mg/ml) at 37°C for 30 min[36]. Whole spleens were mashed through a 70-µm cell strainer to obtain single-cell suspensions. Whole livers were mashed through a 70-µm cell strainer and centrifuged at 55 x g for 3 min to remove hepatocytes. Bone marrow from left and right hind limbs were isolated as described previously[37]. Leukocytes isolated from each organ were further incubated with ACK (ammonium-chloride-potassium) solution for red blood cell lysis except for the erythrocyte progenitor cell detection. Prior to staining with cell surface markers, cells were treated with an Fc receptor block (1 µg/1 × 106 cells; BD Bioscience, San Diego, CA) for the reduction of nonspecific binding. Prior to staining, Zombie Green™ or Zombie UV™ Fixable Viability Kits were used to discriminate between dead and live cells according to instructions of the manufacturer (BioLegend, San Diego, CA). Single immune cells were stained with antibodies conjugated with the indicated fluorochromes for 20 min on ice, including anti-CD3-Alexa488, anti-CD11c-Alexa488, anti-NKG2D-PE, anti-PD-L1-PE/Dazzle 594, anti-Ly6G-PE/Cy7, anti-PD-1-APC, anti-F4/80-APC/Cy7, anti-SiglecF-APC/Cy7, anti-Ly6C-BV421, anti-CD45-BV510, anti-CD11b-BV605, anti-CD4-BV650, anti-MHCII-BV650, anti-CD8-BV785, and anti-CD3-BV785 (BioLegend, San Diego, CA). After washing, the stained cell samples were run on the CytoFLEX 13-color cytometer (Beckman Coulter, Brea, CA, USA) and quantified using the CytExpert analysis software. Immune cell populations were characterized as follows: PMN-MDSC (CD11b+/Ly6G+), eosinophils (CD11b+/F4/80+/SiglecF+), alveolar macrophages (MHCII−/CD11b−/SiglecF+), interstitial macrophages (MHCII+/CD11b+/F4/80+), splenic macrophages (MHCII+/CD11b−/F4/80+), M-MDSC (CD11b+/Ly6C++), natural killer cells (CD3−/F4/80−/NKG2D+), dendritic cells (MHCII+/CD11c+), CD4+ T cells (CD3+/CD11b−/CD4+), CD8+ T cells (CD3+/CD11b−/CD8+), natural killer T cells (CD3+/CD11b−/Nkp46+), and B cells (MHCII+/CD11b−/CD11c−). Each population of immune cells was delineated by different colors: PMN-MDSC (brown), eosinophils (yellow), M-MDSC (light blue), dendritic cells (pink), interstitial macrophages (blue), alveolar macrophages (orange), NK cells (green), B cells (grey). CD4+ T cells (light red), CD8+ T cells (red).
Cytokine detection
Murine blood samples were collected in tubes containing 5 mM EDTA. Plasma was further obtained with 1,000 x g at room temperature for 20 min. Cytokine concentrations in the murine plasma were measured using the LEGENDplex™ Mouse Inflammation Panel (13-plex; BioLegend) according to the manufacturer’s recommendations. Levels of GM-CSF, IL-6, TPO, M-CSF, IL-5, IL-34, CXCL12, EPO, SCF, LIF, IL-3, TGF-β1, IL-15 were analyzed using the CytoFLEX 13-color cytometer (Beckman Coulter, Brea, CA, USA) and BioLegend LEGENDplex software (BioLegend, San Diego, CA).
Western blot analysis
Tissue homogenization was performed by using the Miltenyi gentleMACS dissociator
instrumentation (GentleMACS, Miltenyi Biotec). Proteins were lysed in RIPA buffer (Millpore # 20–188) and quantified by Assay Dye Reagent Concentrate (Bio-Rad #5000006). Gel electrophoresis was separated on sodium dodecyl sulfate–polyacrylamide gel electrophoresis (SDS-PAGE) by using Mini Gel Tank (Invitrogen™, cat. #A25977, Waltham, MA) and gels were transferred to a polyvinyl difluoride membrane (PVDF, 0.45 mm; Immobilon, Millipore, Eschborn, Germany) using the Mini Blot Module (Invitrogen™, cat. #B1000, Waltham, MA). Membranes were further blocked in blocking buffer (3% BSA, 0.1% Tween-20 in PBS, pH 7.4) for 1 hour at room temperature. The primary antibodies against EPO (Sion #59099-T42), ARGINASE 1 (Cell Signaling # 93668T), GRANZYME B (Cell Signaling #44153), TUBULIN (Elabscience #E-AB-20036), and ACTIN (Millipore #MAB1501) were diluted in the blocking buffer and incubated overnight at 4°C with gentle agitation. Membranes were washed with PBST (0.1% Tween 20), and probed with goat anti-mouse (Elabscience # E-AB-1001) or anti-rabbit secondary antibodies (Elabscience # E-AB-1003). Chemiluminescent detection was performed using ECL Plus reagent (PerkinElmer Life Sciences) and detected using a ChemiDoc XRS imaging system (Bio-Rad, Hercules, CA, USA).
Real-time reverse transcription polymerase chain reaction
Total RNA was extracted using TRIzol (Invitrogen) and quantified by Synergy HTX (BioTek). Two µg of total RNA were reversed to cDNA using TOOLS Easy Fast RT Kit (BioTOOLS #KRT-BA06-2) with T100 Thermal Cycler (BIO-RAD). Real-time polymerase chain reaction was performed with ChamQ Universal SYBR qPCR Master Mix (Vazyme #Q711) using CFX Connect Real-Time PCR Detection System (BIO-RAD). The amplification of cDNA was performed under the following conditions: 3 min at 94°C followed by cycles of 15 s/94°C, 20 s/60°C, 20 s/72°C. Primer sequences for the genes of interest including Arg1, Cd8, Ccl2, B2m, Hprt were shown in Table 1. Transcripts for B2m and Hprt served as reference genes.
Table 1
Forward Primer
|
Sequences
|
Reverse Primer
|
Sequences
|
Arg1-F
|
5’-GCCTATCTTACAGAGAAGGT-3’
|
Arg1-R
|
5’-GTCCCTGGCTTATGGTTAC-3’
|
Cd8-F
|
5’-CAGAGACCAGAAGATTGTCG-3’
|
Cd8-R
|
5’-TGATCAAGGACAGCAGAAGG-3’
|
Ccl2-F
|
5’-GCTCAGCCAGATGCAGTTAA-3’
|
Ccl2-R
|
5’-TCTTGAGCTTGGTGACAAAAACT-3’
|
B2m-F
|
5’-ACAGTTCCACCCGCCTCACATT-3’
|
B2m-R
|
5’-TAGAAAGACCAGTCCTTGCTGAAG-3’
|
Hprt-F
|
5’-CTGGTGAAAAGGACCTCTCGAAG-3’
|
Hprt-R
|
5’-CCAGTTTCACTAATGACACAAACG-3’
|
Statistical analysis
Results were expressed as the mean ± standard error of the mean (SEM). Statistical comparisons were analyzed by two-tailed Student t-test or one-way ANOVA. Differences were considered significant at *P < 0.05, **P < 0.01, ***P < 0.001. The graphs were generated and analyzed using GraphPad Prism (Version 9.0 GraphPad Software, San Diego, CA).