Reagents
Biocompatible PEG-PLGA (50:50, MW: 12,000 Da) was obtained from Ruixi BiotechCo., Ltd. (Xian, China). IR780 iodide, G418 and B27 were purchased from Sigma-Aldrich (St. Louis, MO, USA). Dichloromethane (CH2Cl2) was purchased from Beijing Bailingwei Technology Corp. (China). MitoTrackerTM Deep Red FM (MitoTracker) and C-11 BODIPYTM 581/591 (C11-BODIPY) were purchased from Thermo Fisher Corp (USA). Foetal bovine serum (FBS) and Dulbecco’s-modified Eagle’s medium (DMEM) were purchased from HyClone (Logan, UT, USA). The cell viability and cytotoxicity test kit, namely, Cell Counting Kit-8 (CCK-8), Liperfluo and FerroOrange were purchased from Dojindo Molecular Technologies (Kimamoto, Japan). The annexin V-FITC/propidium iodide (PI) double-staining test kit was purchased from KeyGen Biotech (Nanjing, China). RIPA lysis buffer, phenylmethanesulfonyl fluoride (PMSF), a bicinchoninic acid (BCA) protein assay kit, Membrane and Cytosol Protein Extraction Kit (cat. P0033), bovine serum albumin (BSA), 1,1′-dioctadecyl-3,3,3′,3′-tetramethylindocarbocyanine perchlorate (DiI), a JC-1 mitochondrial membrane potential (MMP) assay kit (cat. C2006), a Membrane and Cytosol Protein Extraction Kit (cat. P0033), 2,7-dichlorodihydrofluorescein-diacetate (DCFH-DA) (S0033S), N-acetyl-L-cysteine (NAC) (cat. S0077), an adenosine triphosphate (ATP) assay kit (cat. S0026) and mouse anti-human GAPDH (cat. AG019-1) were obtained from Beyotime Biotech (Shanghai, China). YF633-Phalloidin was sourced from Evrbright, INC (US). Rabbit anti-human Bax (cat. D2E11), rabbit anti-human Bcl-2 (D55G8) (cat. 4223), rabbit anti-human cytochrome c (cat. 11940), rabbit anti-human cleaved caspase-7 (cat. 8438), rabbit anti-human cleaved caspase-9 (cat. 20750), rabbit anti-human cleaved caspase-3 (cat. 9664), and a ferroptosis Antibody Sampler Kit (cat. 29650) were purchased from Cell Signaling Technology (Boston, MA, USA). Z-VAD-FMK (cat. S7023), necrostatin-1 (Nec-1) (cat. S8037), bafilomycin A1 (Baf-A1) (cat. S1413), ferrostatin-1 (Fer-1) (cat. S7243) and deferoxamine (DFO) (cat. S5742) were sourced from Selleck (Houston, USA). PCNA (cat. 10205-2-AP) and Ki67 (cat. 27309-1-AP) were obtained from Proteintech (USA). Epidermal growth factor (EGF), basic fibroblast growth factor (bFGF) was obtained from PeproTech (USA).
Cell culture
The human OS cell lines HOS, MG63, and 143B, the murine OS cell line K7M2, murine breast cancer line 4T1 cells, and human non-small-cell lung cancer A549 cells were purchased from the American Type Culture Collection (ATCC, Manassas, VA, USA) and maintained in DMEM supplemented with 10% FBS, 100 μg/mL penicillin and 100 μg/mL streptomycin. Cells were maintained in a humidified atmosphere with 5% CO2 at 37 °C. The human fetal osteoblastic cell line HFOB 1.19 was also obtained from the ATCC, and the cells were cultured in Dulbecco's modified Eagle’s Medium F-12 nutrient mixture (DMEM/F12) supplemented with 2.5 mM of l-glutamine, 0.3 mg/mL of G418 in addition to 10% FBS, 100 μg/mL penicillin and 100 μg/mL streptomycin. HFOB 1.19 cells were maintained in a humidified 5% CO2 atmosphere at a permissive temperature of 34 °C.
Synthesis of the MH-PEG-PLGA-IR780 NPs
i) The water-in-oil-in-water (W/O/W) double-emulsion method was used to synthesize PEG-PLGA-IR780 NPs. Briefly, PEG-PLGA (50 mg) and IR780 (2 mg) were dissolved in CH2Cl2, and then the primary emulsification was emulsified using an ultrasonic probe (Sonics & Materials, Inc., USA) (50 W, 3 minutes). Subsequently, 4% polyvinyl alcohol (PVA) (10 mL) was added to the emulsified solution, which was homogenized by a second sonication to form a W/O/W double emulsion (35 W, 3 minutes). Then, 2% isopropanol solution (10 mL) was added to the prepared emulsion which was mechanically stirred for 2 h to remove the CH2Cl2 followed by centrifugation (12,000 rpm for 6 minutes) to obtain the PEG-PLGA-IR780 NPs. ii) To prepare the HOS cell membranes, HOS cells were grown in T-75 culture flasks to full confluence and detached with 2 mM EDTA in phosphate-buffered saline (PBS), and washed three times in PBS by centrifuging at 800 rpm for 5 minutes. Subsequently, the cells were lysed with Membrane and Cytosol Protein Extraction Kit A containing PMSF (100:1), at -80°C with repeated freeze-thaw cycles followed by centrifugation (800 rpm for 5 minutes) to collect the cellular supernatant, finally, the precipitate after centrifugation at 12,000 rpm for 30 minutes, was the HOS cell membrane (MH). iii) MH-PEG-PLGA-IR780 NPs were finally synthesized by physically extruding the mixture of PEG-PLGA-IR780 NPs and the MH at the same concentration for 11 passes through a 400 nm polycarbonate porous membrane on a mini extruder (Avanti Polar Lipids, USA). The final product was stored at 4 °C for later use.
Characterization of the MH-PEG-PLGA-IR780 NPs
The size distribution, and zeta-potentials of various NPs and their stability of the NPs in fetal blood serum (10%) and PBS over seven days were determined using dynamic light scattering (DLS, Malvern Instruments, UK). The morphologies and structures of the NPs were observed using transmission electron microscopy (TEM, Hitachi H-7600, Japan). The optical absorption of the different NPs and free IR780 was determined with an ultraviolet-visible (UV-vis) spectrophotometer (US-2550, Shimadzu, Japan). A standard concentration curve of free IR780 measured at a wavelength of 798 nm was constructed to calculate the amount of IR780 encapsulated into the MH-PEG-PLGA NPs. The encapsulation efficiency (EE) and encapsulation content (EC) of IR780 were calculated by Equations (1) and (2)
(1) EEIR780 (%) = (mass of total IR780 - mass of unentrapped IR780/mass of total IR780) ×100%
(2) ECIR780 (%) = (mass of total IR780 - mass of unentrapped IR780/mass of total PEG-PLGA NPs) ×100%
Cellular uptake and deep penetration capability of the MH-PEG-PLGA-IR780 NPs
Laser confocal scanning microscopic (CLSM; Nikon A1+R, Japan) and Flow Cytometry (FC; BD FACSvantage SE, USA) were used to detect the cellular uptake of different NPs. Typically, HOS cells (1×105/dish) were seeded into a laser confocal cell-culture dish. After 24 h of incubation, the culture medium was replaced with the serum-free medium containing PEG-PLGA NPs, PEG-PLGA-IR780 NPs or MH-PEG-PLGA-IR780 NPs (stained with DiI; λ excitation/λ emission = 549nm/565nm) for 1, 2, 3 or 4 h (IR780, 20 μg/mL). Then, the cells were fixed in 4% formaldehyde for 10 minutes and washed with PBS. After incubation for different time intervals with various NPs, the cell nuclei were stained blue with DAPI (λ excitation/λ emission = 364 nm/454 nm). The fluorescence images were directly recorded by CLSM. Moreover, the quantitative cellular uptake of PEG-PLGA NPs, PEG-PLGA-IR780 NPs and MH-PEG-PLGA-IR780 NPs at different time intervals was quantified and analyzed by FC.
The penetration of the MH-PEG-PLGA-IR780 NPs was estimated in vitro using 3D tumor spheroid models. HOS tumor spheres (1×105/well) were cultured in 6-well ultra-low attachment plates (Corning, Tewksbury, MA) in stem cell medium that consisted of serum-free DMEM/F12 with 20 ng/mL EGF, 20 ng/mL bFGF and B27. The medium was changed every two days over ten days. Then, the medium was replaced by the DiI-labelled NPs dispersed in DMEM/F12 (1 mL, 20 μg/mL). After 4 h of coincubation, the 3D tumor spheroids were stained with DAPI for 10 minutes, and observed using CLSM.
Mitochondrial location and macrophage uptake assay
To verify the mitochondrial location of the MH-PEG-PLGA-IR780 NPs, MitoTracker was used to the label mitochondria. Typically, HOS cells (1×105/dish) were seeded into a laser confocal cell- culture dish for 24 h of incubation. Before the test, the growth medium was replaced with DiI labeled NPs (PEG-PLGA NPs, PEG-PLGA-IR780 NPs or MH-PEG-PLGA-IR780 NPs) at a concentration of 20 μg/mL for a 4 h of coincubation. Then, the cells were fixed in 4% formaldehyde for 10 minutes and washed with PBS. Nest, the HOS cell mitochondria were labelled with MitoTracker (λ excitation/λ emission = 644 nm/665 nm) for 30 minutes, followed by 10 minutes of staining with DAPI. Finally, the mitochondrial localization of the NPs was observed using CLSM, and the Pearson correlation (PC) coefficients of each image were measured. This same staining method was used for 4T1, A549, MG63, 143B, K7M2 and HFOB 1.19 cells to identify the homologous targeting capabilities of the MH-PEG-PLGA-IR780 NPs.
Next, to verify the immune escape ability of the MH-PEG-PLGA-IR780 NPs, RAW 264.7 cells (1×105/dish) were seeded in a laser confocal cell- culture dish. After a 24 h of incubation, the growth medium was replaced with serum-free medium containing DiI labeled NPs (20 μg/mL) for 4 h of coincubation. Subsequently, the cells were fixed in 4% formaldehyde for 10 minutes and washed with PBS, YF633-Phalloidin and DAPI were used to label the cytoskeleton and nuclei of the RAW 264.7 cells within 10 minutes and 30 minutes, respectively. The uptake of various NPs by macrophages was finally observed by CLSM.
FL imaging/biodistribution and PA imaging of the MH-PEG-PLGA-IR780 NPs In vivo
HOS tumor-bearing mice were intravenously injected with MH-PEG-PLGA-IR780 NPs suspension (2 mg/mL, 200 μL). Subsequently, NIR FL images were collected, pre-injection and at 1, 2, 3 and 6 h post-injection, and the relative FL intensity of each tumor region was measured by IndiGo 2.0.5.0 (Berthold Technologies, Germany). Ex vivo imaging was performed on the major organs and tumor tissues at 6 h post-injection to detect the biological distribution of the MH-PEG-PLGA-IR780 NPs.
A Vevo LAZR Photoacoustic Imaging System (VisualSonics Inc., Toronto, Canada) was used to determine the PA performance of the MH-PEG-PLGA-IR780 NPs. It has been reported that the PA signal intensity of IR780 is the greatest at the activation wavelength of 798 nm [27]. HOS tumor-bearing mice were intravenously injected with a MH-PEG-PLGA-IR780 NPs suspension (2 mg/mL, 200 μL). PA images were collected at different time points (pre-injection, 1, 2, 3 and 6 h post-injection), and the corresponding PA signal intensities were measured with a Vevo LAZR System.
Cell viability assay
A CCK-8 assay was used to determine cell viability after the different treatments. Briefly, HOS cells were seeded into 96-well plates at a density of 5,000 cells/well and incubated overnight for adherence. To determine the safety of the different NPs in vitro, HOS cells were incubated with various concentrations of NPs (NPs: 0.0, 0.2, 0.4, 0.6, 0.8, 1.0 mg/mL, and IR780: 0.0, 6.5, 13.0, 19.5, 26.0, 32.5 μg/mL) for different length of time (0, 12, 24 and 48 h). To assess the change in cell viability after pretreatment with different inhibitors (z-VAD-FMK, Nec-1, Baf-A1, DFO, Fer-1, NAC), HOS cells were pre-incubated with the aforementioned inhibitors for 24 h before exposure to MH-PEG-PLGA-IR780 NPs-associated PDT. Following different treatments, 10 μL of CCK-8 to added to each well followed by incubation for an additional 1 h. Finally, the plates with cells were placed in a microplate reader (MK3, Thermo Scientific) to measure the absorbance at 450 nm. Cell viability was calculated using the following formula: cell viability (%) = experimental group absorbance value/control group absorbance value×100%.
Measurement of intracellular ROS
Intracellular ROS was detected using DCFH-DA (λ excitation/λ emission = 488 nm/530 nm). Typically, HOS cells (1×105/dish) were seeded into a laser confocal cell- culture dish. After a 24 h of incubation, the medium was replaced by serum-free medium with or without NPs (PEG-PLGA-IR780 NPs or MH-PEG-PLGA-IR780 NPs (IR780: 20 μg/mL)) followed by incubation for another 4 h. Then, the cells in the laser group received irradiation with an 808 nm laser at a power density of 1.5 W/cm2 for 2 minutes. Then, the cells were incubated in serum-free medium containing 10 μM DCFH-DA in the dark at 37°C for 30 minutes. Next, the cells were washed with PBS to remove excess DCFH-DA. Finally, the cells were immediately observed by CLSM to detect the intracellular ROS levels, treated with trypsin, collected in 200 µL PBS and detected by FC.
MMP assay
The MMP (Δψm) was detected using a JC-1 assay kit. HOS cells (1×105/dish) were seeded into a laser confocal cell- culture dish for a 24 h of incubation. After adherence, the HOS cells were divided into different treatment groups (control, laser alone, PEG-PLGA-IR780 NPs, laser+PEG-PLGA-IR780 NPs, MH-PEG-PLGA-IR780 NPs, and laser+MH-PEG-PLGA-IR780 NPs (IR780: 20 μg/mL)). After 4 h of intervention, all cells were cultured in fresh medium containing 1 mL of JC-1 staining solution (10 μM) and incubated for 30 minutes in the dark at 37 °C under 5% CO2. Then, the cells were washed with ice-cold JC-1 buffer, and JC-1 aggregates (λ excitation/λ emission = 585 nm/590 nm) and JC-1 monomers (λ excitation/λ emission = 514 nm/529 nm) were observed by CLSM. To quantify the MMP, after exposure to different treatments, the cells were trypsinized, collected in medium containing JC-1 staining solution (10 μM), and incubated in the same environment for 30 minutes. After washing with ice-cold JC-1 buffer, the cells were suspended in 200 µL of PBS for FC analysis.
TEM
TEM was used to detect the changes in mitochondrial morphology after different treatments to assess ferroptosis induction. HOS cells were seeded overnight into a 6-well plate at a density of 105 cells/well to allow adherence and then divided into groups for various different treatments (control, laser alone, PEG-PLGA-IR780 NPs, laser+PEG-PLGA-IR780 NPs, MH-PEG-PLGA-IR780 NPs, and laser+MH-PEG-PLGA-IR780 NPs (IR780: 20 μg/mL)). Next, the cells were trypsinized, collected, and fixed with 2.5% glutaraldehyde and 1% osmic acid. Then, the cells were dehydrated with a graded ethanol series and acetone, embedded, sliced, and stained with 3% uranyl acetate-lead citrate. Finally, the cells were examined by TEM (JEM-1400 Plus, JEOL, Japan).
LPO and Lipid-ROS measurements
To determine the levels of LPOs and Lipid-ROS, HOS cells (1×105/dish) were seeded into a laser confocal cell- culture dish for 24 h of incubation. Then, the cells were treated differently (control, laser alone, PEG-PLGA-IR780 NPs, laser+PEG-PLGA-IR780 NPs, MH-PEG-PLGA-IR780 NPs, laser+MH-PEG-PLGA-IR780 NPs (IR780: 20 μg/mL)). Subsequently, the cells were stained with 5 μM LPO (λ excitation /λ emission = 534 nm/535 nm) or 2 μM C11-BODIPY (λ excitation/λ emission = 500 nm/510 nm) for 30 minutes at 37°C in the dark. Next, the cells were washed with PBS to remove the excess LPO and C11-BODIPY. Finally, CLSM was used to observe LPO and Lipid-ROS levels in the cells, which were then trypsinized and collected for FC analysis.
Fe2+ detection
FerroOrange (λ excitation/λ emission = 561 nm/570 nm) was used to detect the level of intracellular Fe2+. Briefly, HOS cells were seeded into a laser confocal cell- culture dish at a density of 105 cells/well overnight to allow adherence and then treated with laser alone, PEG-PLGA-IR780 NPs, laser+PEG-PLGA-IR780 NPs, MH-PEG-PLGA-IR780 NPs, laser+MH-PEG-PLGA-IR780 NPs (IR780: 20 μg/mL). Then, the cells were stained with 1μM FerroOrange for 30 minutes at 37°C in the dark. Next, the cells were washed with PBS to remove the excess FerroOrange. Finally, the cells were observed by CLSM to detect the intracellular Fe2+ levels, and then treated with trypsin, collected in 200 µL of PBS and detected by FC.
Cell apoptosis assay
Apoptosis was determined by FC using annexin V-FITC/PI staining. Briefly, cells were seeded into 6-well plates (1×105 cells/well) and incubated overnight for adhesion. Then, the cells were exposed to laser alone, PEG-PLGA-IR780 NPs, laser+PEG-PLGA-IR780 NPs, MH-PEG-PLGA-IR780 NPs, or laser+MH-PEG-PLGA-IR780 NPs (IR780: 20 μg/mL). After treatments, the cells were collected, washed twice with ice-cold PBS, and stained with annexin V-FITC and PI according to the manufacturer’s instructions. Finally, the samples were suspended in 200 µL of PBS and then analysed by FC.
Western blot
After HOS cells were treated according to the different regimens, the cells were lysed with RIPA lysis buffer containing PMSF and phosphatase inhibitor within a specified period of time to extract the total protein in the cells. Protein samples (30 to 50 μg/lane) were separated on a 10%-12% gel by SDS-PAGE and transferred to polyvinylidene fluoride (PVDF) membranes, which were blocked with 5% skim milk for 1.5 h at room temperature, and then incubated overnight with the corresponding primary antibody (1:1000) at 4℃. Next, the membranes were washed with Tris-buffered saline with Tween-20 (TBST) and incubated with the secondary antibody (1:8000) at 37°C for 1 h. Finally, the reactive protein bands on the membrane were detected with an enhanced chemiluminescence (ECL) detection system and developed on film.
Intracellular ATP level
An ATP assay kit was used to determine the intracellular ATP levels. Briefly, HOS cells were grown in 6-well plates at a density of 105 cells/well and incubated overnight for adhesion. After the cells were exposed to laser alone, PEG-PLGA-IR780 NPs, laser+PEG-PLGA-IR780 NPs, MH-PEG-PLGA-IR780 NPs, laser+MH-PEG-PLGA-IR780 NPs (IR780: 20 μg/mL), the cells were trypsinized and centrifugation (12,000 rpm for 5 minutes) at 4°C. Finally, the cell supernatant was collected for RLU detection with a luminometer equipped with a multimode reader (260-Bio, Thermo Fisher Scientific, USA).
Xenograft tumor model
Thirty male BALB/c nude mice (4 weeks old) were supplied by the Experimental Animal Center of Chongqing Medical University. All animal studies were approved by the Ethics Committee of Chongqing Medical University. The mice were housed with free access to a commercial diet and water under specific pathogen-free conditions. After the mice were acclimated for 1 week prior to initiation of the study, HOS tumor-bearing mice were established by subcutaneous injection of 200 μL of sterile PBS containing a HOS cell suspension at a density of 106 cells/mL. After the tumor volumes reached 50 mm3, different treatments were initiated. Thirty mice were randomized into the following 6 groups: (1) control, (2) laser only, (3) PEG-PLGA-IR780 NPs, (4) laser+PEG-PLGA-IR780 NPs, (5) MH-PEG-PLGA-IR780 NPs, (6) laser+MH-PEG-PLGA-IR780 NPs (laser power: 2 W/cm2, 5 minutes of irradiation) (PEG-PLGA concentration: 5 mg/mL, volume of 200 µL). The tumor volume sizes and mouse weights were measured every 4 days for 16 days after treatment, with the tumor volume calculated according to the following formula: 1/2×a2b (where a is the short axis and b is the long axis of the tumor). Mice were sacrificed under anaesthesia on day 14, and the xenograft tumors of each animal were weighed and analyzed.
In order to avoid photothermal effect (PTT) and to ensure only effects due to the PDT treatment, the temperature of the tumor region was monitored during irradiation with a Xenogen IVIS Spectrum imaging system (PerkinElmer, USA) so that the temperature was always below 42°C. Experiments in vitro was operated on ice.
Haematoxylin-eosin (H&E) and immunohistochemistry (IHC)
After sacrifice, mouse tissues from the xenograft tumors, hearts, livers, spleens, kidneys and lungs were dissected out and fixed in 10% formalin for histopathological studies. After fixation, the tissues were dehydrated in a graded series of ethanol and xylene, embedded in paraffin, cut into sections, and stained with H&E. The expression of PCNA and Ki67 in the xenograft tumor tissues was detected by IHC. Briefly, the paraffin-embedded specimens were separated, fixed with 4% paraformaldehyde, and embedded in paraffin. After embedding, the specimens were discontinuously cut into 4 mm thick sections with a microslicer. Tumor sections were blocked and immunostained with antibodies targeting Ki67 (1:200) or PCNA (1:200). Finally, images were captured using a microscope, and PCNA and Ki67 expression was evaluated by counting the number of positive cells from 5 randomly selected fields in the residual viable tumor tissue among the necrotic areas under a light microscope at a magnification of 200×. Data are presented as the percentage of positive cells.
Biosafety of the MH-PEG-PLGA-IR780 NPs and PDT
To determine the toxicity of the MH-PEG-PLGA-IR780 NPs, they were injected into BALB/c nude mice (PEG-PLGA concentration: 5 mg/mL, volume: 200 µL) that had been randomly divided into 5 groups. Twenty-five mice were euthanized on days 0, 1, 7, 14, and 28 post-injection. The vital organs were collected for H&E staining, and blood samples were sent for the blood index (routine blood and biochemistry) analyses. In addition, the methods of determining the biosafety of MH-PEG-PLGA-IR780 NPs-mediated PDT and the other parallel groups were consistent with the aforementioned methods after the mice were sacrificed at 16 days post-irradiation.
Statistical analysis
All data are expressed as mean ± SD and were analyzed with SPSS 22.0 software. Single Student’s t-test and one-way ANOVA were used to determine statistical significance between pairs of groups or three or more groups, respectively. Significance levels are shown as *p < 0.05, **p < 0.01, and ***p < 0.001.