Methods
Preparation methods and related characterization
2 mL ferric chloride solution (200 mM) was slowly dripped into 20 mL of freshly prepared OVA aqueous solution (0.025 g/ mL) under gentle stirring at room temperature. Amino acid residues in protein molecules captured and chelated metal ions. Then the pH was regulated to 12 through adding aqueous solution of NaOH (2 M). After the system pH changed, the captured ions undergo formed iron hydroxide in situ with rapid growth. Adjusted the pH value to 4 with 0.5% (W/V) citric acid after 2 hours, the denatured proteins were removed by centrifugation (10000 rpm, 10 min) to obtain [email protected] nanoparticles. Then 2 mL IR820 liquor (1 mg/mL) was instilled to the remaining mixture and guided IR820 to coat on the surface of [email protected] nanoparticles through electrostatic adsorption. After stirring for 24 hours, [email protected] nanovaccine was formed. The removal of unloaded free IR820 was carried out by centrifugation and ultrafiltration (10 kDa)
The average diameters and ζ-potentials of [email protected] and [email protected] were checked by dynamic light scattering (Zetasizer ZS90, Malvern, UK). Images of nanovaccine were obtained by 120kV transmission electron microscope (TEM) (Talos L120C G2, Thermo Fisher Scientific, USA).
Release of iron ions was tested by a microplate reader (Varioskan Flash, Thermo Fisher Scientific, USA). In detail, 2 mL prepared [email protected] was put into dialysis bags that could intercept molecular weight at 3500 Da (Yuanye, Shanghai, China). The dialysis bags were divided into two groups and soaked in 30 mL PBS buffer with pH values of 5.5 and 7.4, and kept shaking at 100 rpm in water bath at 37℃. At every time points of the time curve, 1 mL PBS solution was extracted as planned, then the absorbance was measured under 550 nm UV light (SP-756P, Shanghai Spectral Instrument Co., LTD). The concentration of released iron ions was calculated by using the method of iron colorimetric assay kit (Applygen, Beijing, China). After each extraction 1 mL isothermal medium was added into container timely.
Encapsulation efficiency (EE) and drug loading (DL) of [email protected]
The amounts of Fe and IR820 added to the system during the preparation of nanovaccine were recorded as WFe−1 and WIR820−1. Recorded the weight of prepared samples as W1 after lyophilization. Then nanoparticles of a certain quality (W2) was measured and dissolved, then the OVA content was detected by BCA kit (Beyotime, Shanghai, China). Ultraviolet absorption scanned spectrogram of solution to acquire OD value, which was converted to calculate concentration of IR820 to obtain OVA weight (WOVA) and IR820 weight (WIR820) severally. The weight of Fe(OH)3 was measured by WFe(OH)3= W2- WOVA - WIR820. Ultimately, the EE and DL of each substance can be calculated:
$$\text{EE\% of }{\text{Fe}\left(\text{OH}\right)}_{\text{3}}\text{ }\text{=}\frac{{\text{W}}_{{\text{Fe}\left(\text{OH}\right)}_{\text{3}}}\text{×}\frac{{\text{W}}_{\text{1}}}{{\text{W}}_{\text{2}}}\text{×}\frac{{\text{Mr}}_{\text{Fe}}}{{\text{Mr}}_{{\text{Fe}\left(\text{OH}\right)}_{\text{3}}}}}{{\text{W}}_{\text{Fe-1}}}\text{×}\text{100}\text{\%}$$
1
$$\text{EE\% of IR820=}\frac{{\text{W}}_{\text{IR820}}\text{×}\frac{{\text{W}}_{\text{1}}}{{\text{W}}_{\text{2}}}}{{\text{W}}_{\text{IR820-1}}}\text{×100\%}$$
2
$$\text{DL\% of }{\text{Fe(OH)}}_{\text{3}}\text{=}\frac{{\text{W}}_{{\text{Fe(OH)}}_{\text{3}}}}{{\text{W}}_{\text{2}}}\text{×100\%}$$
3
$$\text{DL\% of IR820=}\frac{{\text{W}}_{\text{IR820}}}{{\text{W}}_{\text{2}}}\text{×100\%}$$
4
Evaluation of photothermal performance
[email protected] nanovaccine and free IR820 were dissolved at gradient concentrations (5, 10, 20, 50 µg/mL, calculated by IR820) and irradiated with a power density of 2.5 W/cm2 (808 nm, 10 min), using near-infrared laser emitted from infrared fiber laser (Leoptics, Shenzhen, China). Infrared thermal imager (Fotric 225) was employed to collect temperature of the solution.
Detection of glutathione consumption and proficiency test of extracellular ⋅OH
Under the condition of pH 8.0, phthalaldehyde and GSH can react to produce fluorescent substances. 18 mL OVA, [email protected], [email protected] solutions (5 µg/mL, calculated by IR820) was respectively mixed with 1 mL GSH solution (2 mM). Then 1 mL phthalaldehyde ethanol solution (8 mg/mL) was instilled. After blending and incubating for 20 min murkily, fluorescence value of different groups was measured (340 nm excitation and 430 nm emission) by multi-functional microplate analyzer (Tecan Infinite F200, Tecan, Switzerland). The GSH content was described as the ratio of the relative content with the blank group.
In addition, intracellular GSH concentration was determined by GSH and GSSG detection kit (Beyotime, Shanghai, China). B16-OVA cells were incubated with OVA, [email protected], [email protected], [email protected] + NIR (5 µg/mL, calculated by IR820) for 24 h after which procedure the [email protected] + NIR group was irradiated for 5 min (808 nm, 2.5 W/cm2). Without nanoparticles cells were used as control. Cellular glutathione levels were then measured according to product specifications. The relative level of glutathione content in treatment group was calculated by comparing with control group.
Crystal violet can be discolored by hydroxyl radicals generated by decomposition of hydrogen peroxide. This experimental process can be used to verify whether [email protected] nanoparticles can catalyze the formation of hydroxyl radicals. The crystal violet solution was mixed with hydrogen peroxide and [email protected], which was left at room temperature for 1 hour. Crystal violet and hydrogen peroxide as control group and all samples were scanned by UV-vis spectroscopy.
Cellular uptake and distribution
B16-OVA cells were inoculated for one day to make adherence. After, the cell medium was added with [email protected] and free IR820 (5 µg/mL, calculated by IR820) for 2, 4, 8, 12 h. Later rinsed B16-OVA cells with PBS and cultured them with 1 mL DMEM medium containing lysosome probe (50nM) at 37℃ for 30 min. After swilling and immobilization, DAPI was used for staining B16-OVA cells for 15 min, followed by observation of laser scanning confocal microscope (LSCM) (Leica, Wetzlar, Hesse, Germany). "ImageJ" software was used to quantitative test mean intracellular fluorescence intensity.
In vitro cytotoxicity assay
After culturing for one day, the medium of B16-OVA cells (overexpressing OVA) was exchanged to 200 µL [email protected] NPs and IR820 DMEM medium with the following concentrations: 0.1, 0.2, 0.5, 1, 2, 5 µg/mL (calculated by IR820), and then the cells were incubated for another 24h. Irradiated half of the orifice plate for 5 minutes (808nm, 2.5 W /cm2). The relative viability of B16-OVA was assessed through MTT method (n = 5).
To certify the occurrence and effect of ferroptosis, B16-OVA cells were added to blank medium and medium mixed with OVA, [email protected], [email protected] (5 µg/mL, calculated by IR820). Each parallel group was added with different related-pathway inhibitors, including DFO (62.5 µM after mixing), Fer-1 (50 nM after mixing), VE (12.5 µM after mixing) and GSH (2.5 mM after mixing). All groups were incubated for one day and MTT method was used to assess relative viability of B16-OVA (n = 5).
In vitro validation of markers related to ferroptosis pathway induced by nanovaccine
B16-OVA cells were exposed blank medium, OVA, [email protected], [email protected] (5 µg/mL, calculated by IR82) for 6 h after seeded into 6-well plates for one day. Irradiated half of holes containing [email protected] 5 minutes (808 nm, 2.5 W/cm2). DCFH-DA (Beyotime, Shanghai, China) staining method was applied to detect intracellular ROS levels and BODIPY581/591-C11 (Thermo, Waltham, MA) could be used as an LPO sensor to detect intracellular LPO level[34]. The B16-OVA cells were stained with ROS sensor (10 µM) after the cell fragments were cleaned by PBS to detect intracellular ROS production. The dye could be changed to LPO sensor (5 µM) and incubated for 20 min to detect LPO production. After staining and washing, B16-OVA cells were observed under LSCM.
Analyzation of GPX4 and detection of intracellular H2O2 level
Western blotting was chosen to evaluate GPX4 levels. B16-OVA cells were cleaved on ice with NP40 lysis buffer containing 0.1% protease inhibitor cocktail after treated with different groups (5 µg/mL, calculated by IR820) and the above methods. The cytoplasmic proteins collected by centrifugation after lysis were boiled to degenerate, followed by electrophoresis in 12% SDS-PAGE Bis-Tris Gel (Invitrogen, Carlsbad, CA), and then transferred onto nitrocellulose membranes with 0.22 µm pores. 5% skim milk powder was prepared to soak and wash the membranes. Then membranes were sequentially incubated with primary antibodies of β-actin (Beyotime, Shanghai, China) or GPX4 (Absin, Shanghai, China) and corresponding fluorescent secondary antibodies. Images were collected by chemiluminescence method and analyzed by "ImageJ" software.
Micro hydrogen peroxide detection kit (Solarbio, Beijing, China) was employed to detect. After incubating with nanovaccines for 24 h, and half of holes containing [email protected] were irradiated for 5 minutes (808 nm, 2.5 W/cm2). Blank medium cells without nanoparticles were added as control. B16-OVA cells were gathered and put into ultrasonic cell breaker to crush. After operation, transferred treated cells and measured the absorbance value at 415 nm.
Detection of immunogenic cell death intracellular biomarkers
ATP detection kit (Beyotime, Shanghai, China) was used to measure ATP content in cells. B16-OVA cells were incubated [email protected], [email protected] and ferrostatin-1. Cells without nanoparticles treatment were added as control. Half of holes containing [email protected] were irradiated for 5 minutes (808 nm, 2.5 W/cm2). Lysate was added at the ratio of one-tenth of the amount of cell culture medium, and lysate cells were operated on ice. The RLU value of supernatant collected by centrifuge was detected by luminomete. In order to eliminate the effect of protein concentration, the intracellular total protein amount was determined to more accurately display the released ATP concentration.
B16-OVA cells were exposed to OVA、[email protected] and [email protected] –IR820 (5 µg/mL, calculated by IR820)for 12 h, and half of the Wells (808 nm, 2.5 W /cm2) containing [email protected] were irradiated for 5 minutes. The control group was added blank cell medium without nanoparticles. After treatment, 4% paraformaldehyde was added for cell immobilization. Followed by cleaning with PBS again, corresponding primary antibody of calreticulin (Beyotime, Shanghai, China) and high-mobility group box 1 (Absin, Shanghai, China) were added and placed overnight at 4℃. After staining with Alexa 488 labeled secondary antibody and washing, B16-OVA cells were observed under LSCM.
Detection of nanovaccine-uptake by dendritic cells
After the C57BL/6 mice (6-8weeks) were sacrificed, removed their femur and tibia in a sterile environment, and rinsed with 75% alcohol, PBS and RPMI 1640 full medium in sequence. Washed the bone marrow cavity with 1–2 mL full medium repeatedly for each bone to collect cells. After centrifugation of the obtained bone marrow cells to collect and lyse the red blood cells in the bone marrow, they were collected through a 74 um filter membrane. The BMDCs were seeded on a 10 mm sterile petri dish. The culture suspension was gently sucked to evenly mix cells when reached the sixth day, and the collection was centrifuged to discard the supernatant. After suspending and incubating with IR820 and [email protected] for 4 hours, flow cytometry (LSRFortessa, BD, USA) and confocal microscopy were used to evaluate the entry of nanovaccine into dendritic cells.
Induction of dendritic cell maturation in vitro
After 7 days of culturing, dendritic cells were transferred from a 10 mm sterile petri dish to a sterile 6-well plate. B16-OVA cells were inoculated with [email protected] and [email protected] + ammonium glycyrrhizinate (0.2 mg/mL) for 12 h respectively. Half of the holes added with [email protected] were irradiated with 808 nm laser (2.5 W/cm2) for 5 min, and cultured to collect damaged debris, which were then added to the BMDCs in the 6-well plate.
Dendritic cells after 7 days of culturing were exposed to 5 µg/mL (calculated by IR820) OVA, [email protected], [email protected] for 24 h. Dendritic cells were dyed by fluorescent antibodies against CD11c, CD80 and CD86. Flow cytometry (Becton, Franklin, USA) was used detect and analyze.
Anti-tumor experimental model construction of C57BL/6 mice
When B16-OVA cells grew to 2.5×105 cells/well, they were collected with 100µL PBS solution and transplanted subcutaneously (s.c.) into the right back of male C57BL/6 mice aged 6–8 weeks to establish a melanoma model as the primary tumor. Four days after the initial inoculation, a second tumor was seeded contralateral as an artificial analog of metastasis. All laboratory mice were randomly divided into 6 groups (n = 5) :(1) Control; (2) OVA; (3) [email protected]; (4) [email protected]; (5) [email protected] + NIR; (6) Fe @ OVA - IR820 + NIR + CTLA-4. Three days later, when the primary tumors of B16 grown on C57BL/6 mice had reached about 200 mm3, 50µL of the nanoagent was intratumoral injected into these tumors (~ 5 µg/mL, calculated by IR820). Two hours later, tumors (right) injected with nanovaccines in groups [email protected]、[email protected] + NIR、[email protected] - IR820 + NIR + CTLA-4 were exposed to laser 10 min for radiation therapy (808nm, 2.5 W/cm2) at 3-minute intervals of 2 minutes. PBS injection group was the control group. This point in time was defined as day 0. Gave the medicine every two days. On days 2, 5, 8, 11 and 14, anti-CTLA-4 antibody was intraperitoneally (i.p.) injected at a dose of 2.0 mg/kg. The weight and tumor volume of each mouse were noted during therapy. Tumor volume could be estimated by the following equation:
$$\text{Tumor volume=}\frac{\text{Length×}{\text{Width}}^{\text{2}}}{\text{2}}$$
5
The mice were euthanized after two weeks. Paraffin was chosen to embed extracted tumors and organs for hematoxylin and eosin (H&E) staining. The observation was under upright microscope (BX63, Olympus, Japan). The lungs of different treatment groups were soaked in Bouin's solution for 24 hours to record pulmonary metastasis of melanoma.
Enhanced antitumor immune responses
Tumor was dehydrated with sucrose and were prepared to develop frozen sections. To explore changes in immune level induced by vaccine treatment in vivo, the content of calreticulin (CRT) and high-mobility group box 1 (HMGB1) in tumor tissues were monitored by immunofluorescence staining. Besides, lymph nodes of mice were removed and the maturation of DCs was analyzed with comparation. Tumor, spleen and lymph node were removed from the mouse cadaver, ground with 70mm filter and washed. Followed instructions of animal tumor infiltrating tissue lymphocyte separation fluid kit (Solarbio, Beijing, China) to separate and purify the lymphocytes in the tumor tissue. Fluorescein - coupled antibodies were used to dye individual cells. The staining regimen for dendritic cells was the same as in vitro to make dyed cell suspension. Intratumoral infiltration of CD8 + T cells and splenic CD4/CD8 + T cells was detected and compared by flow cytometry. Besides, purified lymphocytes were collected from the tumor to detect and analyze the NK cell activation in the tumor. The collected cells were dyed with CD3e and NKp46 antibody (Becton, Franklin, USA) then measured by flow cytometry. All samples were analyzed using FlowJo software.
The excised tumor tissue was treated with ultrasonic cell fragmentation instrumen to obtain individual tumor cells. The levels of interferon γ (IFN-γ), tumor necrosis factor α (TNF-α) and interleukin 6 (IL-6) were measured by enzyme-linked immunosorbent assay (ELISA). Tumor samples of each group were prepared according to instructions and determined according to ELISA kit protocol.