Peptide synthesis.
The LHRH analogue containing a dK residue (d for D configuration of the Lys residue) in position 6 (peptide A: pEHWSY(dK)LRG-amide) and DRS-B2 (peptide B:
GLWSKIKEVGKEAAKAAAKAAGKAALGACSEAV-acid) were synthesized by X'PROCHEM Company (Lille, France). The chimeric peptide H-B2 (Fig. 1A) composed of the peptide A grafted by its epsilon NH2 of the dK6 residue with the COOH-terminal of the peptide B is determined to be >95% pure by RP-HPLC (Fig. 1B) and its molecular weight determined by ESI-mass spectrometry is [M+4H]/4=1105.2 (Fig. 1C). The peptide is dissolved in sterile water for experimental use.
Circular dichroism (CD) spectroscopy of peptides.
The helical structure of HB2 and DRS-B2 were analyzed by CD spectroscopy using a Jobin Yvon CD6 dichrograph linked to a PC micro-processor as described 31. Briefly, measurements were calibrated with (+)-10-camphorsulfonic acid and performed with 10 mM HB2 or DRS-B2 diluted in PBS alone or with increasing concentrations of dodecylphosphocholin (DPC) (10, 30, 100, 1000 and 5000 μM) at 25 ̊C using a quartz cuvette (Hellma) with a path length of 0.1 cm. Spectra, recorded in 1 nm steps, were averaged over five scans, and corrected for the base line. The CD spectra were deconvoluted using CDNN Software 32. Circular dichroism measurements are reported as Δε/n, where Δε is the dichroic increment (M-1 cm-1) and n is the number of residues in the peptide. The α-helix content of peptides was obtained using the relation: Pα = -[Δε222nm x10] (Pα: percentage of α-helix; Δε222nm: dichroic increment per residue at 222 nm) 33.
Fluorescence of tryptophan-containing peptides
performed as previously described by Dos Santos et al.5 Emission spectra were recorded on a Jobin-Yvon Fluoromax II instrument (HORIBA Jobin-Yvon, France) equipped with an Ozone-free 150 W xenon lamp. The excitation wavelength was 290 nm, and the emission spectra were acquired at 300–360 nm. At least five measurements for each titration point were recorded with an integration time of 1 s. The HB2 and DRS-B2 concentration in PBS was 2 μM, and the DPC concentration varied from 0 to 1000 μM. Tryptophan fluorescence was determined by subtracting spectra without peptide.
Nuclear Magnetic Resonance (NMR) Spectroscopy.
The NMR sample was prepared in 550 μL of H2O/D2O (90:10 v/v) in the presence of 85 mM DPC-d38 (d38 -dodecylphosphocholin, MAPCHOâ-12-d38, Avanti Polar Lipids) and using a peptide concentration of 1 mM. The pH was set to 4.1 using microliter amounts of NaOH 0.1 M. DSS was added at a concentration of 0.11 mM for chemical shift calibration. The NMR spectra were acquired at 45°C on a 500 MHz (11.7 T) Avance III Brukerâ spectrometer equipped with a TCI cryoprobe. The following experiments were recorded: 2D 1H-1H TOCSY (66 ms mixing time), 2D 1H-1H NOESY (150 ms mixing time), 2D natural abundance 15N-1H and 2D 13C-1H HSQC experiments, as described 31.
Cell culture.
Human PCa cell lines PC3, DU145 and 22Rv1 were purchased from ATCC (American Type Culture Collection and were cultured in RPMI supplemented with 10% of fetal bovine serum (FBS). Human hyperplasic cell line BPH1 was purchased from the German Collection of Cell Cultures (DSMZ, Braunschweig, Germany). BPH1 were maintained in RPMI 1640 supplemented by 10% FBS, 20 ng/mL testosterone and 1% insulin-transferrin-selenium. Cell cultures were maintained at 37°C and 5% CO2 in humidified atmosphere. All culture reagents were purchased from Life Technologies (Cergy-Pontoise, France). All experiments were performed as previously described by Dos Santos et al.5
Cell-viability assays.
Cells were seeded at a density of 5×103 cells/well in 96-multiwell plates in complete medium and incubated for 24 h at 37˚C in a controlled humidified 7% CO2 environment. Cells were then treated with DRS-B2 or HB2 as indicated, for 48 h. Cell viability was measured using the 3-(4,5-dimethylthiazol2-yl)-diphenyltetrazolium bromide (MTT) dye method (Sigma, Saint Quentin Fallavier, France) according to the manufacturer’s instructions. Each experiment was performed in triplicate and at least three independent experiments as previously described by Dos santos et al.5. IC50 values were determined by GraphPad Prism 5.0 (GraphPad Software, USA).
Small Interference RNA (siRNA) Assay.
PC3 cell silencing of LHRH-R were performed using Lipofectamine RNaiMax (Invitrogen) according to the manufacturer’s protocol as previously described by Elahouel et al.34. Predesigned siRNAs of either non-targeting siRNA (GGCUACGUCCAGGAG CGCACCTT) as a control or target-specific siRNAs (AAGCAUGGAUUGGAUCAGUAATT) to knock-down LHRH-R were obtained from Eurofins Genomics. Briefly, cells were plated overnight at 50-60% confluence, then transfected with 10 nM of either non-targeting siRNA or LHRH-R target-specific siRNAs, in serum- and antibiotic-free Opti-MEM medium (Invitrogen). After 24 h, transfected cells were treated with HB2 as indicated for 24 h. In addition, transfection efficiency was evaluated by Western blot analysis 24-48 h after transfection.
Western blotting
was performed as previously described by Elahouel et al.34 Cell lysates were prepared with RIPA extraction buffer containing anti-protease inhibitors (Roche). Equal amounts of total proteins were separated by SDS-polyacrylamide gel electrophoresis (SDS-PAGE) under reducing conditions, then transferred onto polyvinylidene difluoride membrane (Millipore) using standard protocols. Nonspecific binding was prevented by incubating the membranes in 5% (w/v) nonfat milk in PBS containing 0.2% (v/v) Tween-20 (blocking buffer (BB) for 1 h and then incubated with primary antibodies in diluent (5% nonfat milk in TBS-Tween) for 1 h. Membranes were subsequently incubated overnight at 4°C with antibodies, rabbit anti- GnRH-R (Abcam,183079 ; 1 : 2000) or mouse anti- β-actin (SantaCruz Biotechnology; 1 : 2000) and then with HRP-conjugated secondary antibody for 1h at RT (Cell Signaling Technology ; 1:2000). Immunocomplexes were revealed using a chemiluminescence detection system kit “SuperSignal™ West Pico PLUS” (Thermoscientific) and visualized using the GBox system (Syngene).
Hemolysis assessment.
The hemolytic activity of H-B2 and DRS-B2 was determined using fresh human erythrocytes from a healthy donor that was prepared as follows: 4 mL of whole blood was collected and centrifuged at 900 g for 10 min at 4°C to separate plasma from erythrocytes. The pellet was then rinsed with PBS pH 7.4 and centrifuged at 900 g for 10 min at 4°C. After counting and making a red cell solution at 4 × 108 erythrocytes/mL (diluted in PBS pH 7.4), 50 µL of a diluted peptide solution was added in cascade to which 50 µL of the erythrocyte solution was added (made in triplicate). After 1 hour of incubation at 37°C, the tubes were centrifuged at 12 000 rpm for 15 seconds at 4°C. The supernatant was then recovered. The hemoglobin present in the erythrocytes was determined in the supernatants via a plate reader at 450 nm. A parallel incubation in the presence of 0.2% (v/v) Triton was carried out to determine the absorbance associated with 100% hemolysis.
Tumor xenograft Studies
were performed as previously described by Van Zoggel et al.11. PC3 (2 × 106) cells were injected subcutaneously into the right flank of 4-week-old male NMRI nude mice (Janvier, Le Genest-Saint-Isle, France). When the tumor volume reached approximately 100 mm3, the mice were randomly divided into four groups (n=6): control (PBS), DRS-B2 (2.5 mg/kg), H-B2 (2.5 mg/kg), H-B2 (5 mg/kg) by intraperitoneal injection twice a week. Tumor size was measured two times per week with a caliper and tumor volume was calculated with the formula: V= 4/3π*R12*R2 whereby radius 1 (R1), radius 2 (R2). At the end of the experiment, the mice were sacrificed, and body weight was measured. The tumors were isolated, weighted and then fixed in formalin.
Ethics statement.
All mouse experiments were performed according to ARRIVE guidelines on animal care and approved by Charles Darwin Animal Experimentation Ethics Committee (CEEACD/N°5) and conducted in compliance with European Community.
Immunohistochemical analysis of the tumors
were performed as previously described by Van Zoggel et al.11. The fixed tumors were embedded in paraffin and then 6 µm sections were prepared. The tumors sections were deparaffinized, antigen unmasking was performed, and endogenous peroxidase activity was inactivated with a 2% hydrogen peroxide solution for 10 minutes. Unspecific staining was blocked using Power Block Universal reagent (Biogenex Laboratories/Microm Microtech, Francheville, France) for 10 min at 37°C. Tissues were then incubated 2 hours at room temperature with anti-human Ki67 antibody (Mouse monoclonal, M7240, Dako, 1:50) for 2 hours. Immuno-complexes were revealed using HRP conjugated secondary antibodies and the DAB substrate. Tissues were then counterstained with hematoxylin and cover slipped with Mowiol mounting medium. Quantification of Ki67 positive stained cells was quantified by image J software analysis on the whole tumor section.
Lactate dehydrogenase (LDH) release assay.
The cytoplasm LDH release assay was performed as previously described 11. Briefly, PC3 cells were grown in a 96 well plate (1,500 cells/well/100 μL) in complete medium and treated with various concentrations of H-B2 or DRS-B2. Cell membrane integrity was evaluated by measuring the LDH activity released into the culture media 24 hours after peptide exposure. The CytoTox96 non-radioactive cyto- toxicity assay (Promega; Charbonnières-les-Bains, France) was performed according to the manufacturer’s instructions and quantified by measuring the absorbance at 490 nm. The 100% cytotoxicity corresponds to the LDH released with treatment of the cells with Triton X100 at 0.9% (v/v).
Apoptosis analysis by flow cytometry.
For the apoptosis assay, an FITC-Annexin-V (A-V) and Propidium Iodide (PI) double staining method was used. PC3 cells were grown in a 12-well plate and treated or not with H-B2, DRS-B2 or hydrogen peroxide as described previously 5. 24 hours after exposure, cell viability was evaluated by flow cytometry analysis with FITC-Annexin-V (A-V) and propidium iodide staining. Medium and trypsinized cells were collected and washed with PBS. After centrifugation, cells were suspended in PBS to obtain a cell density of 0.5 × 106 cells per mL. One mL of this cell extract was centrifuged, suspended in 200 μL PBS, transferred to a microtiterplate with round bottom and centrifuged again. Resulted cell pellet was resuspended in 200 μL of Binding Buffer 1x (BD PharmingenTM), containing 5 μL of FITC-Annexin-V (BD PharmingenTM) and incubated during 10 minutes in dark at room temperature. The cells were washed with PBS and incubated with 200 μL of Binding Buffer 1x containing PI (final concentration 1 μg/mL) (BD Pharmin-genTM)) for 5 minutes in dark at room temperature. Flow cytometry analysis was performed with LSR Fortessa X20 analyzer (BD Biosciences) and FlowJo V10 software.
Terminal transferase-mediated dUTP Nick End-Labelling (TUNEL) assay.
PC3 cells were cultured in 6-well plates at 150,000 cells per well in complete medium and treated or not with 1 or 2.5 µM of DRS-B2 or H-B2 and with LHRH (5 mM) or Taxotere (10 nM) and Staurosporin (1 mM) as positive control. After 24 hours of treatment, the supernatant was aspirated and the cells trypsinized and plated on a Superfrost PLUS slide and fixed with a 4% formaldehyde solution without methanol. The cells were then permeabilized with a 0.2% Triton X-100 solution in 1X PBS for 5 minutes at room temperature. Terminal transferase-mediated dUTP nick end-labelling (TUNEL) was performed according to the DeadEnd™ Fluorometric TUNEL System kit from Promega®. In addition to the fluorescein labeling, a second labeling with DAPI, intercalating DNA was performed by incubating for 15 minutes at room temperature with a 1 µg/mL solution. The different labelling steps were performed avoiding any exposure to light.
Imaging was performed with an Axiolmager M2 epifluorescence microscope. The images obtained by microscopy were recorded with the Zen 2012 software and analyzed with the ImageJ© software. Each image underwent a thresholding of fluorescence calibrated on the positive control, and which was preserved for all other images. The software then analyzed the number of DAPI and fluorescein fluorescence events to calculate the ratio between the total number of DAPI-labeled cell nuclei and the number of fluorescein-labeled nuclei with fragmented DNA. The experiment was performed in triplicate.
Statistical analysis.
The statistical analyses were performed using the GraphPad PrismTM version 4.00 software from GraphPad Software Inc. (San Diego, CA, USA). The results are expressed as the means ± standard deviation (SD) or standard error mean (SEM) of at least three determinations for each test from three independent experiments. Statistical analyses were carried out using the unpaired t-test. The statistical significance of the differences is given as *p < 0.05; ** p < 0.01; ***p < 0.001; ns: not significant.