MATERIALS AND METHODS
Reagents
Except where otherwise indicated, reagents were purchased from Sigma-Aldrich Corporation (St. Louis, MO, US), and reactions were performed at room temperature (22°C–24°C). The tested compounds were synthesized as previously described (
11,
51–53).
Data acquisition and bioinformatics analysis
The reference
L. infantum proteome (JPCM5, proteome ID UP000008153) was downloaded from UniProt [release 2022_01 (
20)]. The subcellular location of
L. infantum proteins was extracted from UniProt annotations and manually cured. Transmembrane annotated proteins were analyzed with DeepTMHMM v1.0.15, an improved version of TMHMM 2.0 [
https://dtu.biolib.com/DeepTMHMM/ (
54)], capable of identifying transmembrane beta-barrels. Propensity to protein aggregation was assessed using the Aggrescan prediction method (
21), and the Normalized a4vSS for 100 residues (Na4vSS) was acquired as the aggregation reporter. Sequences over 6000 residues were divided into smaller fragments, and the Na4vSS for the whole ensemble was calculated. For the
L. infantum full proteome, the mean aggregation propensity is displayed. The content in prion-like proteins was assessed with the PLAAC algorithm (
18) using the whole proteome for each case as the background probability, and those proteins with at least one window with COREscore > 0 were considered prion-like. Structures for
L. infantum tubulin β chain (A0A381MS01) and cleavage and polyadenylation specificity factor-like protein (A4I7V5) were obtained from the AlphaFold database (
30) version 3 and colored with Pymol. Amyloid prediction was carried out with Waltz (
29) in its default “best overall performance” mode. GO enrichment analysis was performed with the functional annotation tool of the Database for Annotation, Visualization and Integrated Discovery knowledge base update v2022q3 (
22), where GO terms annotated directly by the source (GO_DIRECT category) were selected and the
L. infantum JPCM5 data set was chosen as background.
Peptide synthesis
Acetyl-DNFIFGQ-amide and acetyl-AISVFFLEP-amide were synthesized at a 0.1-mmol scale on H-Rink amide-ChemMatrix resin (PCAS BioMatrix Inc., Saint-Jean-sur-Richelieu, QC, Canada) in a Prelude (Gyros Protein Technologies AB, Uppsala, Sweden) synthesizer running Fmoc solid-phase peptide synthesis (SPPS) protocols. After chain assembly, the N-terminus was deprotected and acetylated (acetic anhydride/N,N-diisopropylethylamine, 1 mmol each, 45 min, dimethylformamide), prior to full deprotection and cleavage with trifluoroacetic acid (TFA)/H2O/triisopropyl-silane (95:2.5:2.5 vol/vol), 90 min, at room temperature. Peptides were precipitated from the TFA solution by the addition of cold diethyl ether followed by centrifugation (3 × 4800 rpm, 5 min, 4°C), taken up in water, and lyophilized. Crude peptides were checked by analytical reverse-phase high-performance liquid chromatography (RP-HPLC) and liquid chromatography-mass spectrometry (LC-MS) and purified by preparative RP-HPLC. Analytical RP-HPLC was performed on a LC-20AD instrument (Shimadzu Corporation, Kyoto, Japan) equipped with a Luna C18 column (3 µm, 4.6 mm × 50 mm; Phenomenex, Torrance, CA, US) using linear gradients of solvent B [0.036% TFA in acetonitrile (ACN)] into A (0.045% TFA in H2O) over 15 min, at a flow rate of 1 mL/min and UV detection at 220 nm. Preparative RP-HPLC was performed on a LC-8 instrument (Shimadzu Corporation) fitted with a Luna C18 column (10 µm, 21.2 mm × 250 mm; Phenomenex), using linear gradients of solvent D (0.1% TFA in ACN) into C (0.1% TFA in H2O) over 30 min, with a flow rate of 25 mL/min. MS analysis was performed on a LC-MS 2010EV instrument (Shimadzu Corporation) fitted with an XBridge C18 column (3.5 µm, 4.6 mm × 150 mm; Waters, Cerdanyola del Vallès, Spain), eluting with linear gradients of F [0.08% formic acid (FA) in ACN] into E (0.1% FA in H2O) over 15 min at a 1-mL/min flow rate. Fractions with the expected mass and >95% purity by LC-MS were pooled and lyophilized. Peptide stock solutions were prepared in sterile deionized water and stored at ‒20°C. Fluorescent versions of the peptides were prepared by coupling 5(6)-carboxyfluorescein to the N-terminus of the peptide resins, as described for acetylation. The fluorescein(Flu)-labeled Flu-DNFIFGQ-amide and Flu-AISVFFLEP-amide were obtained after resin cleavage, preparative RP-HPLC purification, and LC-MS characterization as above.
In vitro peptide aggregation assays
To completely disaggregate the peptides before the assay, around 15 mg of each lyophilized peptide was dissolved in 1 mL of TFA. After thoroughly mixing, TFA was evaporated under a N2 stream and 0.5 mL of 1,1,1,3,3,3-hexafluoro-2-propanol (HFIP) was added, mixed well, and evaporated as before (repeated twice to fully remove TFA). Then, 2 mL of HFIP was added, 40-nM peptide aliquots were prepared in low-binding Eppendorf tubes (Eppendorf, Hamburg, Germany), and HFIP was evaporated overnight in a desiccator. For aggregation assays, peptide stocks were prepared in PBS at a final concentration of 200 µM peptide and incubated at 37°C under stirring at 1,400 rpm in a ThermoMixer (Eppendorf) for 24 h (t24h). Then, in a 96-well flat-bottom black plate (Greiner Bio-One, Frickenhausen, Germany), the peptide solutions were diluted in PBS and ThT was added to obtain final concentrations of 180 µM peptide and 25 µM ThT. The fluorescence emission of ThT was collected from 460 to 800 nm using an excitation wavelength of 450 nm (Infinite Nano+ multimode microplate reader; Tecan Trading AG, Männedorf, Switzerland). As a non-aggregation control, the ThT fluorescence of a freshly disaggregated 180 µM peptide solution (t0) was measured.
Transmission electron microscopy
Peptides (25 µM) were incubated in PBS at 37°C for 24 h under orbital stirring at 300 rpm. After 5 min of water bath sonication (FB15053 ultrasonic bath; Thermo Fisher Scientific Inc., Waltham, MA, US), a 5-µL drop of the peptide solution was deposited onto a carbon-coated copper grid (Ted Pella, Redding, CA, US). Next, 5 µL of 2% uranyl acetate was added and left for 5 min before washing it away with H2O. The resulting samples were observed with a JEM 1010 transmission electron microscope (JEOL Ltd., Tokyo, Japan). Images were acquired using an Orius 832 CCD camera (Gatan Inc., Pleasanton, CA, US).
Encapsulation of YAT2150 in liposomes
Liposomes were prepared by the thin lipid film hydration method (
55). Briefly, the lipids 1,2-dioleoyl-
sn-glycero-3-phosphocholine (DOPC):1,2-distearoyl-
sn-glycero-3-phosphoethanolamine-
N-[maleimide(polyethylene glycol)−2000] (DSPE-PEG-Mal):cholesterol (75:5:20 molar ratio) were mixed in chloroform:methanol (2:1 vol/vol) in a glass vial together with 100 µM YAT2150 from a YAT2150 stock solution in DMSO. Organic solvents were removed by evaporation under nitrogen. The thin lipid film formed on the vial walls was hydrated in PBS, in a volume corresponding to a final lipid concentration of 10 mM. Then, three rounds of constant vortexing for 2 min followed by bath sonication at 35°C for 3 min (FB15053 ultrasonic bath) were performed. Liposomes were extruded through 200 nm polycarbonate membranes (Avanti Polar Lipids Inc., Alabaster, AL, US) using a mini extruder device (Avanti Polar Lipids Inc.). Non-encapsulated YAT2150 was removed by pelleting liposomes by ultracentrifugation (150,000 ×
g, 1 h, 4°C) and replacing the supernatant by PBS, except in the case of cytotoxicity assays, where the pellet was taken up in Medium 199 (M-199) supplemented with 1% penicillin-streptomycin. Fluorescein-labeled liposomes containing a molar ratio of 0.5% CF-PE were prepared as described above, except for a reduced DOPC content of 74.5%. The average diameter and polydispersity index of liposomes were measured after 1:100 sample dilution in PBS, using a Zetasizer NanoZS90 (Malvern Ltd., Malvern, UK). To determine the drug content, liposomes were disrupted by treatment with one volume of 4% SDS, and YAT2150 was quantified by absorbance spectroscopy at a wavelength of 490 nm (Epoch microplate spectrophotometer; BioTek Instruments Inc., Winooski, VT, US). Encapsulation efficiencies are defined as the fraction of YAT2150 in the liposome pool, after ultracentrifugation, relative to the initial amount of drug added to the liposomes. The amount of YAT2150 in the sample is expressed relative to the total lipid concentration of 10 mM, assuming that all the lipids end up in liposomes.
Immunoliposome preparation and characterization
Immunoliposomes were prepared by attaching the anti-lipophosphoglycan (LPG) monoclonal IgM antibody CA7AE (Bio-Rad, Hercules, CA, US) to the maleimide group of DSPE-PEG-Mal following established protocols (
49). The
N-succinimidyl
S-acetylthioacetate crosslinker (SATA; Thermo Fisher Scientific Inc.) was first coupled to the antibody at a molar ratio of 1:10 (1 mg antibody/mL in 350 µL of PBS) through an incubation step of 30 min at room temperature. Non-reacted SATA was removed by buffer exchange. Thioester groups from SATA-anti-LPG antibody were deacetylated with 0.1 volumes of deacetylation solution (0.5 M hydroxylamine, 25 mM EDTA in PBS) and incubated for 2 h at room temperature. Then, another buffer exchange was performed with PBS containing 10 mM EDTA to remove the remaining deacetylation solution. To 100 µL (10 mM total lipid in PBS) of a suspension of liposomes exposing maleimide groups and carrying YAT2150, were added 55 µL of thiolated SATA-anti-LPG antibody and 45 µL of PBS supplemented with 10 mM EDTA. The resulting sample was incubated for 17 h at room temperature, when unbound SATA-anti-LPG antibody and YAT2150 were removed by ultracentrifugation (150,000 ×
g, 1 h, 4°C) and replaced with PBS containing 10 mM EDTA. The YAT2150 concentration in immunoliposomes was quantified as specified above.
The presence of anti-LPG antibody and YAT2150 in liposomes was assessed by SDS-PAGE. Ten microliters of each sample was mixed with 10 µL of 2× Laemmli sample buffer (0.125 M tris base, 4% SDS, 20% glycerol, 10% 2-mercaptoethanol, and 2 mg/mL bromophenol blue) and heated at 95°C for 5 min. Then, the samples were run in a 12% SDS-PAGE (Mini Protean II System; Bio-Rad), and the gel was stained with silver nitrate following the next steps: fixation (40% ethanol, 10% acetic acid) for 30 min, washing three times with deionized water (Milli-Q system; Millipore Corporation, Burlington, MA, US), staining for 40 min (1 mg/mL silver nitrate, 0.02% formaldehyde), and developing (25 mg/mL Na2CO3, 0.01% formaldehyde) until visualization of the protein bands, when 1% acetic acid was used to stop the reaction. Then, the YAT2150 fluorescent signal was detected in a LAS 4000 reader (ImageQuant TL; GE Healthcare, Chicago, IL, US) with the Cy3 filter, and an image of the protein bands was taken by white epi-digitalization.
Gel-assisted formation of giant unilamellar vesicles
GUVs were produced by the gel-assisted method with minor modifications (
56). A 5% (wt/wt) polyvinyl alcohol (PVA, 145 kDa; Merck, Darmstadt, Germany) solution was prepared in PBS at 90°C and used to coat a microscope coverslip which was later dried for 30 min in an oven at 50°C. Then, 20 µL of the lipid formulation (10 mM total lipid) DOPC:cholesterol:CF-PE (79.5:20:0.5 molar ratio) was spread over the dried PVA. To evaporate the solvent, the glass slide was placed under vacuum for 30 min at room temperature. Two coverslips and a Teflon spacer were assembled forming a chamber (2-mL volume) that was filled with PBS. One hour later, GUVs were harvested and YAT2150 was added at a final concentration of 2.5 µM. Fifteen microliters of the resulting GUV suspension was transferred to a 2% bovine serum albumin (BSA)-coated coverslip and observed with a Leica TCS SP5 laser scanning confocal fluorescence microscope (Leica Microsystems, Wetzlar, Germany). Fluorescein and YAT2150 were detected by excitation at 488 and 561 nm and emission collection in ranges 497–543 and 572–657 nm, respectively.
Cytotoxicity assays
Human umbilical vein endothelial cells were maintained in M-199 supplemented with 10% fetal bovine serum (FBS) and 1% penicillin-streptomycin at 37°C, 5% CO2, and seeded in 96-well plates at a density of 50,000 cells/mL. After allowing cell adherence for 24 h, the medium was removed, and drugs or nanoformulations were added in M-199 supplemented with 1% penicillin-streptomycin. Plates were incubated for 48 h, and then, the medium in each well was replaced by 100 µL of M-199 containing penicillin-streptomycin and 0.00125% resazurin sodium salt (Sigma-Aldrich Corporation). Plates were incubated for 4 h, and resorufin fluorescence emission was measured (λex/em: 535/590 nm) in a Tecan Infinite 200 PRO equipment (Tecan Trading AG).
Solubility screen assay
The stock solutions (10‒2 M) of the assayed compounds were diluted to decreased molarity, from 300 μM to 0.1 µM, in a 384-well transparent plate (Greiner Bio-One, Madrid, Spain) with DMSO:PBS (1:99 vol/vol). After 2 h of incubation at 37°C, the plate was read in a NEPHELOstar Plus nephelometer (BMG LABTECH GmbH, Ortenberg, Germany) at 635 nm. The results were adjusted to a segmented regression to obtain the maximum concentration at which the compounds were soluble.
Human plasma stability assay
Plates (96-well Polypropylene Deep Well Plate; Corning Inc., Somerville, MA, US) containing 100 µL of 5 µM compounds in human plasma (Seralab, Granollers, Spain), pooled from healthy donors and extracted in citrate tubes, were incubated at 37°C for different times (0, 60, 120, and 360 min). Then, 300 µL ACN was added to precipitate plasma protein, and the plate was centrifuged at 4000 × g for 60 min at 4°C. For sample quantification, the resulting supernatant was taken and analyzed by ultra performance liquid chromatography with tandem mass spectrometry (UPLC-MS/MS; ACQUITY Xevo TQD UPLC QSM; Waters) in an ACQUITY BEH C18 column (1.7 µm, 2.1 × 50 mm; Waters). As mobile phase (0.6 mL/min flow rate) was used the following gradient of 0.1% formic acid in water (A) or in ACN (B): 95% A:5% B from 0 to 0.1 min, gradual increase to 100% B from 0.1 to 1 min, maintenance in 100% B from 1 to 2 min, return to 95% A:5% B from 2 to 2.1 min, and maintenance of this ratio from 2.1 to 2.5 min. Compound concentrations were calculated from the MS peak areas.
Human hepatocyte stability assay
A vial of human hepatocytes (100 donor pool HCP100.H15/Lot: 2010012; XenoTech, Kansas City, KS, US) was removed from the liquid nitrogen tank and immersed into a water bath. When only a small ice crystal remained, the cell suspension was transferred to a tube with the preheated suspension medium (OptiThaw hepatocyte medium, K8000; XenoTech). After a brief centrifugation at 100 × g for 5 min, hepatocytes were gently resuspended with incubation medium (OptiIncubate hepatocyte medium, K8400; XenoTech) and counted, and viability was determined using trypan blue staining. One-micromolar compounds were incubated at 37°C and 5% CO2 in the presence of 0.5 × 106 hepatocytes in a 500-µL incubation volume. Aliquots were taken, and reactions were terminated with ACN at each of the six sampling time points (0, 15, 30, 60, 90, and 120 min). The samples were centrifuged, and the remaining compound was determined by UPLC-MS/MS analysis as above. Intrinsic clearance was calculated from the logarithm of the remaining compound at each of the times evaluated.
Human microsomal stability assay
The following components were added in a 96-well microplate in a final volume of 500 µL 50 mM Na/K phosphate buffer, pH 7.4 (stock solutions are indicated in parentheses, dissolved in the same buffer unless otherwise specified): 1 µM NADP (10 mM), 10 µM glucose 6-phosphate (100 mM), 1 µM glucose 6-phosphate dehydrogenase (40 U/mL, dissolved in 5 mM sodium citrate), 3 µM MgCl2·6 H2O (30 mM, dissolved in H2O), 0.8 mg protein/mL of human microsomes (10 mg/mL protein; XenoTech), and 1 µM of test compound (100 µM, dissolved in ACN). Plates were incubated at 37°C and 75 µL samples were taken at 0, 10, 20, 40, and 60 min, to which 75 µL ACN + internal standard (Rolipram) to inactivate the microsomes and 30 µL of 0.5% formic acid in H2O to improve the chromatographic conditions were added, and kept at 4°C. When all the samples were taken, the plate was centrifuged at 46,000 × g for 30 min at 15°C, and UPLC-MS/MS was performed as above, except for the time in 100% B from 1 to 2.5 min, the return to 95% A:5% B from 2.5 to 2.6 min, and the maintenance of this ratio from 2.6 to 3.0 min. Metabolic stability was calculated from the logarithm of the remaining compound at each of the times evaluated.
Cytochrome inhibition activity assay
To screen the inhibition potential of the compounds using recombinant human cytochrome P450 enzymes (CYP1A2, CYP2C9, CYP2C19, and CYP2D6) and probe substrates with fluorescent detection, incubations were conducted in a 200-µL volume in 96-well microtiter plates (Costar #3915; Corning Inc.). Addition of cofactor-buffer mixture (0.2 M KH2PO4 buffer, 1.3 mM NADP, 3.3 mM MgCl2, 3.3 mM glucose-6-phosphate, and 0.4 U/mL glucose-6-phosphate dehydrogenase), previously diluted control Supersomes (insect cell control for Supersomes enzymes, baculovirus-insect cell-expressed; Corning Inc.), standard inhibitors (furafylline, tranylcypromine, sulfaphenazole, and quinidine), and test compounds was carried out by a Zephyr liquid handling station (Caliper Life Sciences, Hopkinton, MA, US). The plate was then pre-incubated at 37°C for 5 min, and the reaction was initiated by the addition of pre-warmed enzyme/substrate mix, which contained buffer (KH2PO4), cDNA-expressed P450 in insect cell microsomes, substrate (3-cyano-7-ethoxycoumarin for CYP1A2 and CYP2C19, 7-methoxy-4-(trifluoromethyl)coumarin for CYP2C9, and 3-[2-(N,N-diethyl-N-methylammonium)ethyl]-7-methoxy-4-methylcoumarin for CYP2D6) to obtain the final assay concentrations in a reaction volume of 200 µL. Reactions were terminated after a specific time for each cytochrome by the addition of stop solution (ACN:0.5 M tris-HCl, 80:20 vol/vol). Fluorescence in each well was measured using a fluorescence plate reader (EnVision 2104 multilabel plate reader; PerkinElmer, Waltham, MA, US), and percentage of inhibition was calculated.
Caco-2 permeability assay
Caco-2 cells (ATCC under the license of Abcam Inc., doing business as NaviCyte Scientific, Berkeley, CA) cultured to confluency were trypsinized and seeded onto a filter transwell insert (high-throughput screening Transwell 96-well permeable supports; Corning Inc.) at a density of ~10,000 cells/well in Dulbecco’s modified Eagle’s medium (DMEM). Confluent cells were subcultured at passages 58–62 and grown in a humidified atmosphere of 5% CO2 at 37°C. Following an overnight attachment period (24 h after seeding), the cell medium was replaced every other day with fresh medium in both the apical and basolateral compartments. The cell monolayers were used for transport studies 21 days post-seeding, when monolayer integrity was checked by measuring the transepithelial electrical resistance (TEER). If TEER values were ≥500 Ω/cm2, the medium was removed and the cells were washed twice with pre-warmed (37°C) Hank’s Balanced Salt Solution (HBSS). Compound stock solutions were made in DMSO and further diluted in HBSS (1% final DMSO concentration). All compounds, including controls (colchicine, E3S), were tested at a final concentration of 5 to 10 µM. For A → B directional transport, the donor working solution was added to the apical compartment (A) and HBSS as receiver working solution to the basolateral compartment (B). For B → A directional transport, the donor working solution was added to B and HBSS as receiver working solution to A. The cells were incubated at 37°C for 120 min with gentle stirring. At the end of the incubation, samples were taken from both donor and receiver compartments and transferred into 384-well plates, and compound concentrations were determined by UPLC-MS/MS analysis as for the plasma stability assay. After the assay, Lucifer Yellow (LY) was used to further validate the cell monolayer integrity; cells were incubated with 10 µM LY in HBSS for 1 h at 37°C, obtaining apparent permeability (Papp) values for LY of ≤10 nm/s, which confirmed the well-established Caco-2 monolayer.
L. infantum cultures
L. infantum (MHOM/ES/2016/CATB101) promastigotes were maintained at 26°C in complete Schneider’s medium (Schneider’s insect medium supplemented with 10% FBS, 25 µg/mL gentamycin, 1% penicillin-streptomycin, and 1% sterile human urine, pH 6.7). For growth inhibition assays, promastigotes were used in the logarithmic growth phase, whereas for the infection of macrophages and generation of amastigotes they were used in the stationary phase. Axenic amastigotes were obtained following established protocols (
40,
57–59); briefly, 1 × 10
7 promastigotes/mL in stationary phase were cultured in Schneider’s insect medium supplemented with 20% FBS, 25 µg/mL gentamycin, and 3.9 g/L of 2-(
N-morpholino)ethanesulfonic acid, at pH 5.4 and 37°C. After 48 h, the typical rounded morphology of axenic amastigotes and their flagellum shortening could be observed. This culture was used within a week after its preparation.
Protein aggregation assays in L. infantum cultures
Ten milliliters of L. infantum promastigote cultures in the logarithmic growth phase at a concentration of 107 cells/mL in T-25 flasks (SPL Life Sciences, Pochon, Kyonggi-do, South Korea) was either treated with 0.15 µM or 0.3 µM YAT2150 or left untreated. After 90 min and 4 h of incubation, 1 mL of each culture was washed (3×, 1 mL PBS, 600 × g, 3 min) and the parasite-containing pellets were taken up in 100 µL of 4.5 mg/mL NaCl supplemented with 1× cOmplete protease inhibitor cocktail (Roche, Basel, Switzerland) and incubated overnight at 4°C with gentle stirring. After this time, samples were spun down (2,000 × g, 10 s) and the protein in each supernatant was quantified with the Pierce BCA protein assay kit (Thermo Electron Corporation, Waltham, MA, US). In a 96-well flat bottom black plate (Greiner Bio-One), 2 µg of protein from each supernatant was diluted in a final volume of 100 µL PBS in duplicates. Protein aggregation was measured by the addition of 25 µM ThT to each well, and after a 15-min incubation with gentle stirring, ThT fluorescence emission intensity was measured as detailed above.
Isolation of aggregation-prone proteins from L. infantum cultures
Proteins insoluble in 0.1% SDS were isolated following previously described protocols (
60). First, 40 mL of a
L. infantum preparation containing approximately 10
8 L. infantum promastigotes in the logarithmic growth phase was spun down (50 ×
g, 3 min) to remove dead parasites and washed twice with sterile PBS supplemented with one tablet of cOmplete protease inhibitor cocktail in 10 mL. After centrifugation (600 ×
g, 3 min) to pellet the parasites, they were taken up in 300 µL of RIPA buffer (150 mM NaCl, 1% Triton X-100, 0.1% SDS, 2 mM EDTA, 5% glycerol, 50 mM tris-HCl, pH 9.4) supplemented with 1× cOmplete. The solution was homogenized in a bath sonicator (FB15053 ultrasonic bath) for six cycles (pulse: 30 s on, 30 s off) and incubated for 90 min at 4°C. Next, the lysate was spun (300 ×
g, 2.5 min, 4°C) to remove debris and unbroken cells and the supernatant was carefully loaded on top of 1 mL of 40% sucrose and ultracentrifuged (200,000 ×
g, 1 h) in order to pellet large insoluble aggregates. These were resuspended in 400 µL of lysis buffer (PBS containing 2% SDS, 5 mM DTT, and 2 mM EDTA; supplemented with 1× cOmplete) and incubated at 37°C for 30 min, pipetting up and down every 2 min with siliconized pipette tips. The resulting sample was spun down (16,000 ×
g, 12 min), and the supernatant was recovered and concentrated using an Amicon ultra 0.5 mL centrifugal filter, 3 kDa cutoff (Sigma-Aldrich Corporation). The protein in the concentrated solution was quantified using the Pierce BCA protein assay kit, and 100 µg of protein was loaded in a 12.5% SDS-PAGE stained with Coomassie Brilliant Blue R-250. The material not entering the resolving gel was excised and subjected to liquid chromatography with tandem mass spectrometry (LC-MS/MS) analysis.
LC-MS/MS analysis
Trypsin digestion of proteins in gel slabs was performed in a ProGest automatic digestor (Genomic Solutions Inc., Ann Arbor, MI, US). Each sample was reduced (20 mM DTT in 25 mM NH4HCO3, pH 8.0, 60 min, 60°C), alkylated (55 mM iodoacetamide in 50 mM NH4HCO3, pH 8.0, 30 min, 25°C, protected from light), and digested for 2 h with 80 ng of porcine trypsin (sequencing-grade modified Trypsin Gold; Promega, Madison, WI, US) in 50 mM NH4HCO3, pH 8.0, 37°C. Another aliquot of enzyme was added, and the digestion was allowed to continue overnight under the same conditions. The resulting peptide mixture was extracted from the gel matrix with 5% FA in 50% ACN followed by 100% ACN, cleaned with a C18 tip (PolyLC Inc., Columbia, MD, US) as per manufacturer’s protocol, and finally dried in a SpeedVac concentrator and stored at ‒20°C until LC-MS/MS analysis. To increase peptide amounts, a third digestion with Proteinase K (Sigma-Aldrich Corporation) was performed. To both the extracted tryptic digest and the remaining gel band were added 100 ng Proteinase K in 500 mM NH4HCO3, pH 8.0, and after a 15 min digestion the resulting peptides were extracted from the gel as described above, pooled with the remaining in-solution digestion, dried in a SpeedVac concentrator and stored at ‒20°C until LC-MS/MS analysis.
Mass spectrometry was performed in a NanoAcquity HPLC system (Waters) coupled to an LTQ-OrbitrapVelos mass spectrometer (Thermo Fisher Scientific Inc.). The dried tryptic digests were taken up in 1% FA, and an aliquot was injected into the liquid chromatography system. Peptides were trapped in a Symmetry C18 trap column (5 µm, 180 µm × 20 mm; Waters) and separated in a C18 reverse-phase NanoAcquity UPLC BEH capillary column (130 Å, 1.7 µm, 75 µm × 250 mm; Waters), with a mobile-phase 1% to 40% B gradient in 30 min followed by a 40% to 60% B gradient in 5 min (A: 0.1% FA in water; B: 0.1% FA in ACN) and a flow rate of 250 nL/min. Eluted peptides were ionized in an emitter needle (PicoTip; New Objective Inc., Littleton, MA, US) with an applied spray voltage of 2 KV. A 300–1,600 m/z range of peptide masses was analyzed in a data-dependent mode where a full scan MS was acquired in the Orbitrap with a resolution of 60,000 full width at half maximum at 400 m/z. Within this range, the 15 most abundant peptides (≥500 counts) were selected from each scan and fragmented in the linear ion trap using collision-induced dissociation (38% normalized collision energy) with He as the collision gas. The scan time settings were as follows: full MS: 250 ms (1 microscan) and MSn: 120 ms. Generated *.raw data files were collected with Thermo Xcalibur (v. 2.2).
A database was created by merging all protein entries present in the public UniProt database for L. infantum (uniprot_Linfantum_taxonomy_5671_cont, v25/3/21) with a small database containing laboratory contaminant proteins. The *.raw data files obtained in the LC-MS/MS analyses were used to search with the SequestHT search engine using Thermo Proteome Discover (v1.4.1.14) against the aforementioned database. Both target and decoy databases were searched to obtain a false discovery rate (FDR) and thus estimate the number of incorrect peptide-spectrum matches that exceeded a given threshold, applying preestablished search parameters [enzyme: trypsin (semi); missed cleavage: 2; fixed modifications: carbamidomethyl of cysteine; variable modifications: oxidation of methionine and deamination of asparagine and glutamine; and peptide tolerance: 10 ppm and 0.6 Da for MS and MS/MS spectra, respectively]. To improve the sensitivity of the database search, the semi-supervised learning machine Percolator was used in order to discriminate correct from incorrect peptide spectrum matches. The Percolator assigns a q-value to each spectrum, which is defined as the minimal FDR at which the identification is deemed correct (0.01, strict; 0.05, relaxed). These q-values are estimated using the distribution of scores from decoy database search. Only proteins identified with at least two peptides (FDR ≤ 5%) are reported.
Promastigote growth inhibition assay
In 96-well microtiter plates (Nunclon Delta surface; Thermo Fisher Scientific Inc.), serial dilutions (1:2) of the drugs or peptides were performed, either free or loaded in nanoformulations, in 100 µL of complete Schneider’s medium, to which one volume of 2 × 106 logarithmic growth-phase L. infantum promastigotes/mL in the same buffer was added. After 48 h, resazurin sodium salt was incorporated at a final concentration of 0.00125% and the plates were incubated at 26°C for another 24 h, when fluorescence from resorufin was measured as described above.
Amastigote growth inhibition assay
Compound activity on amastigotes was determined following the parasite rescue and transformation assay (
61). RAW 264.7 macrophages were seeded in 96-well microtiter plates at 10
5 cells/mL of DMEM supplemented with 10% FBS and 1% penicillin-streptomycin (complete DMEM, DMEMc) in a final volume of 200 µL per well. After an overnight incubation (37°C, 5% CO
2) to allow cell adherence, the medium was removed and 10
6 L. infantum promastigotes/mL were added (1:10 macrophage:promastigote ratio) in DMEM supplemented with 2% FBS and 1% penicillin-streptomycin (DMEM 2% FBS). After 24 h, parasites not internalized by macrophages were removed by washing (3×, PBS) and dilutions in DMEM 2% FBS of free or liposome-encapsulated compounds were added. After 48 h, the medium was removed, cells were washed three times with PBS, and 40 µL of 0.05% SDS in complete Schneider’s medium was added; after 40 s, 160 additional µL/well of complete Schneider’s medium was added and plates were incubated at 26°C for another 48 h. Then, resazurin sodium salt was incorporated at a final concentration of 0.00125% and plates were incubated for 24 h more in the same conditions. Afterwards, resorufin fluorescence was measured as above.
Assessment of variation of ATP levels in L. major
The process described in reference (
62) was followed. Briefly,
L. major promastigotes (Friendlin strain) were transfected with the pLEXSY-hyg2.1 expression vector (Jena Bioscience, Jena, Germany) containing a cytoplasmic form of firefly luciferase mutated at its C-terminal tripeptide to prevent its import into the glycosome. Promastigotes were grown in Roswell Park Memorial Institute 1640 medium (RPMI) supplemented with 10% FBS, 2 mM L-glutamine, 1,000 U/mL penicillin/streptomycin (Gibco, Billings, MT, US), and 100 µg/mL hygromycin (InvivoGen, San Diego, CA, US) at 26°C. To assess variation in the intracellular concentration of free ATP, parasites were harvested at the late exponential phase of growth. After two washes with Hanks balanced buffer, parasites were resuspended in the same buffer supplemented with 10 mM D-glucose at 2.2 × 10
7 parasites/mL. After the addition of DMNPE-caged D-luciferin (GoldBio, St. Louis, MO, US) at 50 µM final concentration, the parasite suspension was aliquoted (90 µL/well) into a black 96-microwell plate (Nunc A/S, Roskilde, Denmark), and luminescence readout was monitored in a POLARstar Galaxy microplate reader (BMG LABTECH GmbH) with a luminescence setup. Once the luminescence reached a plateau, 10 µL of a YAT2150 solution at 10× fold its final concentration was added (
t0) and the luminescence value at this point was considered as 100%. DMSO in the parasite suspension was never above 1%. Controls for membrane permeabilization (0.1% Triton X-100) and inhibition of oxidative phosphorylation (1.5 µM 1,4-naphthoquinone) were used as controls, as well as untreated parasites. Experiments were made in duplicate and repeated at least twice.
Flow cytometry
For the analysis of peptide targeting to promastigotes, 2.5 × 106 promastigotes of the L. infantum MHOM/ES/2016/CATB101 strain in the logarithmic growth phase were placed in Eppendorf tubes in a volume of 500 µL of complete Schneider’s medium. Disaggregated Flu-AISVFFLEP-amide and Flu-DNFIFGQ-amide were reconstituted with complete Schneider’s medium and added to the cells in a final concentration of 50 µM. After overnight incubation, promastigotes were stained for 30 min with 4 µg/mL Hoechst 33342, washed with PBS (3×, 600 × g, 3 min), and finally fixed with 3% paraformaldehyde for 20 min. After three more PBS washes, the samples were diluted 1:5 in a final volume of 500 µL PBS in 5 mL polystyrene round-bottom tubes (Corning Inc.) and processed using a 20-parameter standard configuration in a five-laser LSRFortessa flow cytometer (BD Biosciences, San Jose, CA, US). Side- and forward-scatter were used in a logarithmic scale to determine the cell population, acquiring 10,000 events for each sample. Hoechst 33342 and fluorescein were detected by excitation with 350- and 488-nm lasers, and emission was collected with 450/50-BP and 525/50-BP filters, respectively.
For the analysis of YAT2150 targeting, 5 × 106 promastigotes of the L. infantum MHOM/ES/2016/CATB101 strain in the logarithmic growth phase and 5 × 106 axenic amastigotes from the same strain in day 5 of growth were placed in Eppendorf tubes with a final volume of 1 mL of complete Schneider’s medium. YAT2150 dissolved in DMSO was added in a final concentration of 0.38 µM (final DMSO concentration < 0.1%) along with Hoechst 33342 (4 μg/mL final concentration), incubated for 30 min, and washed (PBS, 300 × g, 3 min). Then, parasites were fixed and analyzed by flow cytometry as specified above, substituting fluorescein detection by YAT2150 detection (excitation with a 561-nm laser and emission collection with a 600LP-610/20-BP filter).
For the detection of antibody targeting to promastigotes, 1 × 107 promastigotes of the L. infantum MHOM/ES/2016/CATB101 strain in complete Schneider’s medium were fixed in an Eppendorf tube for 20 min with 3% paraformaldehyde, washed 3× with PBS, and incubated for 1 h at room temperature with the anti-LPG monoclonal IgM antibody CA7AE diluted 1:500 in PBS containing 0.3% BSA. After three PBS washes, the cells were incubated for 1 h with a secondary goat anti-mouse antibody conjugated to Alexa Fluor 488 (AF488; Invitrogen, Waltham, MA, US) diluted 1:200 in PBS containing 2% BSA. After three final PBS washes, flow cytometry analysis was performed as specified above for peptide targeting with fluorescein, except for the acquisition of 30,000 events.
Fluorescence microscopy
All the samples were visualized in an 8-well chamber slide (ibidi GmbH, Gräfelfing, Germany). For staining with the ProteoStat aggresome detection kit (Enzo Life Sciences Inc., Farmingdale, NY, US),
L. infantum promastigotes and axenic amastigotes in days 3 and 5 of growth, respectively, were washed twice with PBS. In PBS, a 1:2,000 ProteoStat dye dilution was then added and after 5 min, 4 µg/mL Hoechst 33342 was incorporated and incubated for a further 10 min. The samples were then washed (3×, PBS) and observed with a Leica TCS SP5 laser scanning confocal fluorescence microscope. ProteoStat and Hoechst 33342 were detected by excitation through 561- and 405-nm lasers, respectively. Emission was collected between 590 and 670 nm for ProteoStat and between 415 and 500 nm for Hoechst 33342. The same protocol was followed for YAT2150 staining of promastigotes and axenic amastigotes, substituting ProteoStat by an incubation in 0.38 µM YAT2150. To quantify Manders’ overlap coefficient (
63), images were analyzed using the Just Another Colocalization Plugin [JACoP (
64)] in the Fiji software (
65). Manders’ coefficient ranges from 0 to 1 showing the pixel percentage that is overlapped, being 0 defined as no colocalization and 1 as total colocalization.
For the analysis of peptide targeting to promastigotes, 200 µL of the samples prepared as described above in the flow cytometry section was placed in an 8-well chamber slide and fluorescence microscopy was conducted in a Zeiss LSM 800 equipment (Zeiss, Oberkochen, Germany) with a 100×/1.4 oil DIC M27 objective. Hoechst 33342 was excited with a 405-nm diode laser, 5 mW, class 3B, and fluorescein with a 488-nm diode laser, 10 mW, class 3B; emission was collected in the 390–460- and 500–700-nm ranges, respectively. A Z-stack image of 30 layers of a promastigote incubated with the Flu-AISVFFLEP-amide peptide was acquired in a Leica TCS SP5 laser scanning confocal fluorescence microscope, where Hoechst 33342 and fluorescein were excited with 405- and 488-nm diode lasers and emission was collected in the 414–474- and 501–562-nm ranges, respectively.
For the targeting analysis in promastigotes of YAT2150 encapsulated in CF-PE-containing liposomes, 107 L. infantum promastigotes/mL were incubated in complete Schneider’s medium for 1 h with 0.38 µM YAT2150 contained in the liposome suspension, during the last 30 min in the presence of 4 µg/mL Hoechst 33342. Cells were then washed three times with PBS, fixed with 3% paraformaldehyde for 20 min, washed again, and visualized with a Leica TCS SPE laser scanning confocal fluorescence microscope. Hoechst 33342, CF-PE, and YAT2150 were excited with 405-, 488-, and 532-nm solid-state lasers and the respective emissions were collected in the 408–521-, 491–564-, and 589–691-nm ranges.
For the analysis of macrophages treated with YAT2150-stained promastigotes, RAW 264.7 macrophages maintained in DMEMc at 37°C and in the presence of 5% CO2 were seeded at 105 cells/mL in DMEMc in an 8-well chamber slide, placing 300 µL per well and allowing cells to adhere overnight. In parallel, 107 L. infantum stationary-phase promastigotes in 1 mL of DMEM 2% FBS were stained with 0.38 µM YAT2150 for 1 h at room temperature and thoroughly washed with DMEM 2% FBS (3×, 600 × g, 3 min). Then, RAW 264.7 cells were washed once with DMEM 2% FBS and 106 YAT2150-stained L. infantum promastigotes/mL were added to each well and incubated for 24 h. Afterwards, 2 µg/mL Hoechst 33342 and 5 µg/mL wheat germ agglutinin-Oregon Green 488 (Thermo Fisher Scientific Inc.) were added simultaneously and incubated for 10 min, and the samples were visualized with a Leica TCS SP5 laser scanning confocal fluorescence microscope. Hoechst 33342, Oregon Green 488, and YAT2150 were excited with 405-, 488-, and 532-nm solid-state lasers and their respective emissions were collected in the 408–521-, 491–537-, and 605–707-nm ranges. For the targeting analysis in RAW 264.7 macrophages exposed to Leishmania of YAT2150 encapsulated in CF-PE-containing liposomes, the cell samples were prepared as described above, except for the use of non-stained promastigotes. Leishmania-exposed macrophages were incubated in DMEM 2% FBS for 3 h with 0.38 µM YAT2150 contained in the liposome suspension, during the last 10 min in the presence of 2 µg/mL Hoechst 33342. Then the cells were washed and observed as described above for promastigote targeting analysis.
For the observation of intracellular amastigotes, 6 × 107 stationary-phase promastigotes were stained in 1 mL of cold PBS with 2.8 µg/mL of CFSE, incubated for 10 min (37°C, 5% CO2), and washed with PBS (3×, 600 × g, 3 min). The final CFSE-stained promastigote pellet was taken up in DMEMc and added to 5 × 105 RAW 264.7 macrophages/mL that had been seeded in DMEMc and allowed to adhere overnight (10:1 promastigote:macrophage ratio). After 4 h of incubation, promastigotes were removed by washing five times with DMEM and cells were incubated for 10 min with 2 µg/mL of Hoechst 33342. Then, 0.38 µM YAT2150 was added, and after 6 h of incubation, the preparations were observed in an IX-51 Olympus (Tokyo, Japan) fluorescence microscope with a 100× objective. The fluorescence of Hoechst 33342, CFSE-stained parasites, and YAT2150 was detected with the fluorescence filter cubes U-MNU2, U-MWIBA3, and U-MWG2, which have excitation filters of 360–370, 460–495, and 534–588 nm and emission filters of 420, 510–550 and 609–683 nm, respectively.
For immunofluorescence microscopy analysis of the binding of anti-LPG monoclonal IgM antibody CA7AE to live promastigotes, the cells were treated as described above for flow cytometry except for the fixation step, which was omitted, and the staining, after removal by washing in complete Schneider’s medium of primary and secondary antibodies, with 4 µg/mL of the DNA dye Hoechst 33342 for 30 min. After three additional washes (600 × g, 3 min), 300 µL of a suspension of stained and washed promastigotes in complete Schneider’s medium was visualized with an IX-51 Olympus microscope as described above, using for AF488 the CFSE settings. For anti-LPG targeting analysis to live Leishmania-exposed and control non-treated macrophages, 5 × 104 RAW 264.7 macrophages/mL were seeded in 300 µL/well of RPMI supplemented with 10% FBS and 1% penicillin-streptomycin in an 8-well chamber slide, and allowed to adhere for 24 h at 37°C in the presence of 5% CO2. Leishmania-treated macrophages were infected by incubation for 24 h in the presence of 5 × 105 promastigotes of the L. infantum MHOM/ES/2016/CATB101 strain. After 3× PBS washes, the cells were prepared for immunocytochemistry analysis as described above for promastigotes, but using 2 µg/mL Hoechst 33342. Confocal fluorescence microscopy was conducted as described above for the analysis of fluorescein-tagged peptides.
Statistical analysis
Unless otherwise stated, experiments were performed in triplicate and the results are expressed as mean values ± standard error of the mean (SEM). The IC50 and CC50 were calculated through non-linear regression analysis using GraphPad Prism 8.4 (GraphPad software, La Jolla, CA, US). Differences between samples were analyzed by one-way analysis of variance using the same software, considering a significant P value ≤0.05. Mean values and SEM were calculated using Microsoft Office Excel version 2306.