Chemicals
Dulbecco’s modified Eagle’s medium (DMEM), phosphate-buffered saline (PBS), fetal bovine serum (FBS), penicillin/streptomycin, l-glutamin, and Trypsin were obtained from Biochrom (Berlin, Germany). Poly-D-lysine hydrobromide, Triton X100, Bovine Serum Albumin, p-phenylenediamine, glycerol and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) were obtained from Sigma-Aldrich Co. (St-Louis, MO, USA). Paraformaldehyde, toluene, 2-isopropanol, methanol and acetonitrile (both HPLC-MS grade), tris(hydroxymethyl)aminomethane and ethylene glycol-bis(β-aminoethyl ether)-N,N,N′,N′-tetraacetic acid, dimethylsulfoxide (DMSO) and trypan blue were used from Carl Roth GmbH + Co. KG (Karlsruhe, Germany).
C60 synthesis
The pristine C60 aqueous colloid solution was prepared by C60 transfer from toluene to water using continuous ultrasound sonication as described by Ritter et al. [7]. The obtained aqueous colloid solution of C60 was characterized by 0.2 mM C60 concentration, 99 % purity, stability, and homogeneity [7, 35].
Matrix assisted laser desorption ionization-time of flight mass spectrometry
An AximaConfidence Matrix Assisted Laser Desorption Ionization-Time of Flight Mass Spectrometry (MALDI-TOF-MS, Shimadzu, Kyoto, Japan) was used to determine the mass of molecular species in C60 colloid solution. Sample (1 μL) was mixed with equal volume of saturated matrix solution (6.5 mM 2,5-dihidrobenzoic acid in 0.1 % trifluoroacetic acid, 50 % acetonitrile) and spotted on a stainless steel target plate and dried. Desorption and ionization were achieved using a 337 nm nitrogen laser. Mass spectra were obtained at maximal laser repetition rate of 50 Hz within a mass range from 0 to 3000 Da. The MALDI-TOFmass spectrometer was calibrated externally using a mixture of standard peptides: Bradykinin fragment 1-7 (757.40 Da), Angiotensin II (human, 1046.54 Da), P14R (synthetic peptide, 1533.86 Da) and ACTH fragment 18-39 (human, 2465.20 Da) from ProteoMass Peptide&Protein MALDI-MS Calibration Kit. To generate representative profiles, a total of 600 laser shots were accumulated and averaged for each sample. MALDI-TOF-MS data processing was performed using the LaunchpadTM v.2.9 Software (Shimadzu, Kyoto, Japan).
Dynamic light scattering
Short ultrasonication (30 sec, 35 kHz) was applied to remove air bubbles. Size distribution of C60 aqueous colloid solution was evaluated with a Zetasizer Nano S equipped with a He-Ne 633 nm laser (Malvern Instruments, UK). Data were recorded at 37 °C in backscattering mode at a scattering angle of 173°. C60, placed in disposable polystyrene cuvettes, was measured 15 times to establish average diameters and intensity distributions. The autocorrelation function of the scattered light intensity was analyzed by the Malvern Zetasizer Software (Malvern Instruments, UK) with the Smoluchowski approximation.
Ultrasound exposure set-up
The water for the ultrasound water bath was previously degassed with the vacuum pump Savant UVS 400A SpeedVac (Thermo Fisher Scientific Inc., Berlin, Germany). For precise positioning of the plates inside the US water bath, especially the distance between transducer and plate, a plate holder was designed in SOLIDWorks (Dassault Systems, Massachusetts, USA) and 3D printed by ViNN:Lab (Technical University of Applied Sciences Wildau, Germany). The position of the plate holder was aligned precisely with the US transducer and marked for identical positioning of the well plate during every experiment. The well plate in that way was positioned in 25 mm from the US transducer. Plates with cells, seeded and treated with C60 according to the type of the assay described below, were prepared for US treatment. To hinder overheating of the plate, every empty well as well as the spaces between the wells on the plate were filled with 100 μL of filtered water. The US treatment was performed with the US generator 68101 coupled with an MH2 transducer, which was mounted on a water bath (Kaijo, Tokyo, Japan). The US transducer itself was a stainless steel transducer plate installed into a polypropylene tank filled with degassed water. The US transducer had an area of 136x81 mm and a frequency in 950 kHz (∼ 1 MHz). The apparatus for the US exposure is shown schematically in Figure 6. The US transducer was driven at 500 W in continuous mode, that correlated to the spatial average, temporal average intensity ISATA of US in 5.4 W/cm2. The temperature of the sample solution was monitored with a digital thermometer. Thus, different locations of a well as well as a space between wells were compared during different US treatment duration. No temperature increase was found for well plate filled with cell culture medium, preincubated at 37 °C and subjected to the US treatment for 60 sec at 500 W, which longer treatment duration a temperature increase was detected, therefore, the ultrasound treatment duration was limited to 60 sec.
Sonoluminescence detection
The experimental setup for sonoluminescence detection consisted of the photomultiplier tube Hamamatsu R928 (Hamamatsu Photonics, Japan), connected with the Oscilloscope Voltcraft 6150c (Conrad Electronic, Germany) and the power supply Thorn EMI PM28B (Thorn Lighting Ltd, United Kingdom). The 24-well plate was used because its wells match the diameter of the photomultiplier window. US bath and plate were filled with degassed distilled water for better sonication and sonoluminescence intensity [55]. A polyfoam holder was used to position the photomultiplier on top of a well of the 24-well plate. The US bath was additionally coated with aluminum foil and measurements were performed in a dark room to shield the photomultiplier tube from any external light. The photomultiplier tube was used to detect light from sonoluminescence. The obtained data are presented in voltage. A peak-to-peak voltage for the entire waveform (Vpp) during 120 sec was chosen as an indicator of sonoluminescence as an index of a full voltage between positive and negative peaks of the detected waveform of voltage from the photomultiplier.
Cell culture
The human cervix adenocarcinoma cell line HeLa (ACC 57) was kindly provided by Dr. Müller (Division of Gastroenterology, Infectiology and Rheumatology, Charité – Universitätsmedizin Berlin, Germany).
Cells were maintained in DMEM, supplemented with 10 % FBS, 1 % penicillin/streptomycin and 2 mM glutamine and cultured in 25 cm2 flasks at 37 °C with 5 % CO2 in a humidified incubator binder (Tuttlingen, Germany). Treatment with Trypsin (1:10 in PBS) was used to detach adherent cells. The number of viable cells was counted upon 0.1 % trypan blue staining with a Roche Cedex XS analyzer (Basel, Switzerland).
Visualization of intracellular C60 accumulation
HeLa cells (105/mL) were seeded in 6-well plates on glass coverslips, previously coated with poly-D-Lysine, and incubated for 24 h. Cells were treated with 20 µM C60 colloid solution for further 24 h. C60 molecules inside cells were visualized with immunofluorescence staining and fluorescence microscopy. Specific fluorescent dyes were used for co-visualization of subcellular compartments such as mitochondria and nuclei – MitoTracker Orange FM (Invitrogen Molecular Probes, Carlsbad, USA) and 4′,6-diamidine-2′-phenylindole dihydrochloride (DAPI, Sigma-Aldrich Co., St-Louis, USA), respectively. For staining of the mitochondria, cells were washed with PBS and stained with the MitoTracker Orange FM for 30 min at 370C. Then, cells were fixed with 4% paraformaldehyde for 15 min in the dark and permeabilized with 0.2 % Triton X100 for 10 min at room temperature and washed again with PBS. Primary monoclonal antibody IgG against C60 (Santa Cruz Biotech Inc., Santa Cruz, USA) and polyclonal antibody against mouse IgG F7506 labeled with fluorescein isothiocyanate (FITC, Sigma-Aldrich Co., St-Louis, USA) were subsequently used according to (Grebinyk et al. 2018). Finally, the coverslips were rinsed with dH2O, incubated with nucleus staining antifade solution (0.6 µM DAPI, 90 mM p-phenylenediamine in glycerol/PBS) for 2 h in the dark and sealed with slides.
Fluorescence microscopy was performed with the Keyence Microscope BZ-9000 BIOREVO (Osaka, Japan) equipped with blue (for DAPI, λex = 377 nm, λem = 447 nm), green (for FITC, λex = 472 nm, λem = 520 nm) and red (for MitoTracker, λex 543 nm, λem = 593 nm) filters with the acquisition Software Keyence BZ-II Viewer (Osaka, Japan). The merged images and single-cell fluorescence intensity profiles were processed with the Keyence BZ-II Analyzer Software (Osaka, Japan).
Quantification of intracellular C60 accumulation
To study the accumulation dynamics we have extracted C60 from the cell homogenate as well as from the mitochondrial fraction and carried out high- performance liquid chromatography-electrospray ionization mass spectrometry (HPLC-ESI-MS, Shimadzu, Kyoto, Japan) analysis as previously established [36].
Briefly, HeLa cells (105/mL) were seeded in 6-well plates from Sarstedt, (Nümbrecht Germany). After 24 h cells were incubated for 0-48 h in the presence of 20 µM C60. Cells were washed with PBS three times, harvested and frozen-thawed in distilled H2O three times and dried at 80 °C under reduced pressure. C60 was extracted to toluene/2-isopropanol (6:1, v/v) via 1 h sonication. After centrifugation (70 min, 20 000 g) the toluene layer was analyzed with HPLC-ESI-MS. Chromatographic separation of C60 was performed using the column Eclipse XDV-C8 (Agilent, Santa Clara, USA) under isocratic elution conditions with a mobile phase of toluene and methanol. Optimized chromatographic conditions and MS parameters were recently published (Grebinyk et al. 2018).
The mitochondrial fraction, obtained according to [56], was used for extraction of C60 as described above, as well as for measurements of protein concentration [57] and succinate-reductase activity [58], used as a mitochondrial marker to testify enrichment and purity of the fraction.
Cell viability
HeLa cells (104/well), cultured in 96-well cell culture plates from Sarstedt (Nümbrecht, Germany) for 24 h, were treated with the 1 % FBS DMEM medium containing 20 µM C60 for 24 h and exposure to the 1 MHz US treatment. The control cells were treated without and with the equal volume of sterile water as a solvent of C60 colloid solution. Cell viability was determined with an MTT reduction assay [59] at 48 h after US treatment. Briefly, cells were incubated for 2 h at 37 °C in the presence of 0.5 mg/mL MTT. The diformazan crystals were dissolved in DMSO and determined at 570 nm with a microplate reader Tecan Infinite M200 Pro (Männedorf, Switzerland).
Cell viability assay was accompanied with the phase contrast microscopy analysis of HeLa cells under the study with the Keyence BZ-9000 BIOREVO (Osaka, Japan).
Caspase 3/7 Activity
HeLa cells were seeded into 96-well plates (104 cells/well) and incubated for 24 h. The cells were treated with 20 µM C60 for 24 h and subjected to US treatment (0, 20, 40, and 60 sec) as described above. Activity of caspases 3/7 was determined at 24 h after ultrasound exposure using the Promega Caspase-Glo® 3/7 Activity assay kit (Madison, USA) according to the manufacturer’s instructions. Briefly, the plates were removed from the incubator and allowed to equilibrate to room temperature for 30 min. After treatment, an equal volume of Caspase-Glo 3/7 reagent containing luminogenic peptide substrate was added followed by gentle mixing with a plate shaker at 300 rpm for 1 min. The plate was then incubated at room temperature for 2 h. The luminescence intensity of the products of caspase 3/7 reaction was measured with the microplate reader Tecan Infinite M200 Pro (Männedorf, Switzerland).
Cell death type differentiation
HeLa cells, seeded in 6-well plates at a cell density of 6×104 cells/well in 1.5 mL of culture medium, were incubated for 24 h, then the medium was replaced with C60-containing medium. After 24 h of incubation with C60 HeLa cells were treated with US as indicated above. At 24 h after US treatment cells were harvested. Apoptosis was detected by Annexin V-fluorescein isothiocyanate/propidium iodide apoptosis detection kit according to the manufacturer’s instructions. Briefly, cells were harvested and washed with binding buffer. After the addition of FITC-conjugated Annexin V cells were incubated for 15 min at room temperature in dark. Cells were washed with Binding buffer and at 10 min after propidium iodide addition were analyzed with the BD FACSJazz™ (BD Biosciences, Singapore). A minimum of 2×104 cells per sample were acquired and analyzed with the BD FACS™ Software (BD Biosciences, Singapore).
On every histogram of flow cytometry four populations of cells are presented according to green (Annexin V-FITC) and red propidium iodide (PI) fluorescence intensities: viable (Annexin V-FITC negative, PI negative), early apoptotic (Annexin V-FITC positive, PI negative), late apoptotic (Annexin V-FITC positive, PI positive) and necrotic (Annexin V-FITC negative, PI positive) cells.
Statistics
All experiments were carried out with a minimum of four replicates. Data analysis was performed with the use of the GraphPad Prism 7 (GraphPad Software Inc., San Diego, California, USA). Paired Student’s t-tests were performed. The significance level was set at p < 0.01.