Isolation of strains and screening for antimicrobial activity
One gram of soil (from Hamilton, Canada) was mixed with 10 ml of PBS and serially diluted before being plated on a soil agar medium. To prepare soil agar, 100 g of soil was mixed with 700 mL of MilliQ water, shaken for several hours, and centrifuged to remove insoluble particles. The supernatant was mixed with 1.5% agar and autoclaved. Soil/agar plates were incubated at 30°C. Fast-growing colonies were picked over the first 3-4 weeks. To isolate slow-growing colonies, plates were wrapped in plastic (tube bag used for petri dish packaging, SARSTEDT) to prevent dehydration and stored at room temperature (RT) for one year. Colonies that appeared were streaked on fresh Tryptone Soy Agar (TSA; Fischer Scientific) plates. From these plates, 80 colonies were isolated and tested for antibacterial activity by the following procedure. Strains were grown in 10 mL of various nutrient media (Tryptone Soy Broth (Fischer Scientific), half-strength Brain Heart Infusion broth (Fischer Scientific), Davis Minimal Broth (Sigma-Aldrich) with 0.5% peptone (Gibco™ Bacto™)), and crude methanolic extracts were prepared by lyophilizing 1 mL of cell-free supernatant at different intervals over the 10 days. Extracts (10 μL) were spotted on cation-adjusted Mueller-Hinton Agar (MHA; BD Difco™) plates containing lawns of the tester strains A. baumannii C0286 and E. coli BW25113ΔtolCΔbamB.
Fermentation and purification of LAR
The Paenibacillus M2 isolate consistently showed bioactivity in tryptic soy broth (TSB) medium against both tester strains. Early attempts to purify LAR from cells grown in TSB were unsuccessful, partly owing to low yield, prompting optimization of fermentation conditions. Replacing glucose with 10 g/L starch and casein enzyme hydrolysate with casamino acid (17 g/L) in TSB generated CSB (Casein Starch Broth) medium. In CSB, LAR production reached 10-15 mg/L. For LAR purification, the Paenibacillus M2 inoculum, prepared overnight in 100 mL CSB, was diluted 100-fold in 1 L of sterile CSB in a 2.8 L Fernbach Flask and incubated with shaking (200 rpm) for 21-23 h at 30°C. Cells were removed by centrifugation and the cell-free supernatant was treated with pre-activated Diaion HP20 resin (5% v/v) for 2-2.5 hours. Resin was washed with 20% MeOH and adsorbed metabolites were eluted with 100% MeOH. The MeOH extract was dried under vacuum, dissolved in water, and fractionated using an SP-sepharose cation-exchange chromatography. The column was pre-equilibrated with 10 mM ammonium acetate buffer (Buffer A; pH 5.0-5.2). Sample pH was also adjusted in a similar range and loaded through a peristaltic pump. The unbound sample was washed with Buffer A, and elution was performed with 0.5 M, 0.75 M, and 1.0 M NaCl in Buffer A, with the 0.75 M fraction containing LAR. This fraction was further purified using a C18 column on a CombiFlash system (Teledyne Inc.), followed by preparative reversed-phase high-pressure liquid chromatography (RP-HPLC) using a C8 column (Eclipse XDB-C8 SemiPrep 9.4x250, 5μM, Agilent Technologies), yielding LAR as a single peak with >95% purity, confirmed by analytical HPLC. Acetonitrile/water containing 0.07% trifluoroacetic acid (TFA) was employed as the mobile phase for both CombiFlash and HPLC systems. The compound was lyophilized to a white fluffy TFA salt and re-lyophilized with diluted HCl to remove TFA.
Structural characterization of LAR
The chemical characterization of LAR was performed using a combination of mass spectrometry, Marfey’s amino acid analysis, and NMR spectroscopy. Mass spectrometry included Liquid Chromatography-Electrospray Ionization-High Resolution Mass Spectrometry (LC-ESI- HRMS or simply LC-MS), Matrix-assisted Laser Desorption/Ionization mass spectrometry (MALDI-MS,) and MALDI-MSMS. LC-ESI-HRMS data were acquired using an Agilent 1290 UPLC separation module and qTOF G6550A mass detector in positive-ion mode. MALDI-MS and MSMS data were obtained on Bruker UltrafleXtreme MALDI TOF/TOF and reflector detector in positive-ion mode. For amino acid analysis, 0.5-1 mg of the compound was hydrolyzed with 6 N HCl at 110°C overnight, derivatized with Marfey’s reagent (Thermo Scientific), and analyzed by LC-ESI-HRMS. NMR analysis was performed with 15 mg of compound dissolved in deuterated D2O, and 1D and 2D spectra were recorded on a Bruker AVIII 700 MHz instrument equipped with a cryoprobe.
Whole genome sequencing and BGC analysis
Genomic DNA from Paenibacillus sp. M2 was isolated using a DNA isolation kit (Promega) and sequenced using Nanopore and Illumina MiSeq. Unicycler was used to perform a hybrid assembly of Illumina and Oxford Nanopore reads35. SPAdes was first used to generate a short-read assembly graph, which was scaffolded with long reads. Multiple rounds of polishing were conducted using Pilon. A single contig was obtained and analyzed using antiSMASH 6.036 to identify the BGC responsible for LAR synthesis.
Identification of lrc-like BGCs and Phylogenetic tree construction
To explore the diversity of lrc-like BGCs in other bacteria, the LrcC amino acid sequence was used as a query in NCBI BLAST, and the hits were manually checked for redundancy. The genomes of potential hits were analyzed manually and were searched for lrc-like BGCs using antiSMASH 7.037. The BGCs were manually curated and analyzed for Lar-like core-peptide sequences. A total of 29 complete BGCs were identified. The LrcC-like cyclases from these BGCs were aligned using the MUSCLE algorithm (default parameters). The aligned sequences were used to construct a maximum-likelihood (ML) phylogenetic tree using MEGA1138, using the WAG substitution model, with a bootstrap value of 100, and keeping other parameters as default. The tree was rooted to an unrelated lasso-cyclase from paeninodin BGC as an outgroup. The BGCs from different bacterial species were aligned using clinker39 tool and inspected manually for the presence of various genes. The amino acid sequences of core peptides from all BGCs were aligned, and the consensus sequence was derived using JalView v. 2.040 with default parameters.
Heterologous expression of LAR
To express LAR in a heterologous host, all the genes in LAR BGC (lrcA to lrcF) were codon optimized for Streptomyces lividans using GenScript’s optimization tool, and the BGC was synthesized as three GBlocks with sequence homology to each other and to the vector, incorporating synthetic promoter and ribosome-binding sequences (RBSs). The fragments were combined and integrated into the plasmid pIJ1025741, digested with NdeI and KpnI, via Gibson assembly42. The assembly mixture was transformed into E. coli TOP10 electrocompetent cells. Plasmids from selected colonies were analyzed via restriction mapping and further validated by sequencing through Oxford Nanopore technology (Plasmidsaurus). A correct plasmid, designated pIJ10257-lrc (Supplementary Data Fig. 10), was transformed into E. coli ET12567 electrocompetent cells, creating the strain ET12567 pIJ10257-lrc.
To move the plasmid into the heterologous Streptomyces lividans host, we performed triparental conjugation using ET12567 pIJ10257-lrc as the donor strain, ET12567 pR9406 as the helper strain, and spores of Streptomyces lividans XF10 (a derivative of S. lividans TK24 strain) as recipients. The E. coli strains were cultured overnight in LB medium with appropriate antibiotics, then subcultured to mid-exponential phase (OD600 1.0-1.2). Cells were harvested, washed, and mixed with activated Streptomyces spores. This mixture was plated on mannitol-soyflour agar (MSA; g/L: mannitol (20), soy flour (20), and agar (20)) plates and incubated at 30°C overnight. The next day, the plates were overlaid with 1 mL of water with nalidixic acid and hygromycin (final concentration in agar plate as 25 µg/ml and 50 µg/ml, respectively) to eliminate E. coli and to select for exconjugants containing the pIJ10257-lrc plasmid. The resulting Streptomyces strain was termed SL-Lar. To delete the lrcF gene, the pIJ10257-lrc was used as a template for PCR amplification using the primers LrcΔF-F1, -F2, -R1, and -R2 (Supplementary Table 1) to yield two fragments. The resulting fragments were assembled and successfully integrated into S. lividans, as described above, to yield S. lividans pIJ10257-lrcΔF (SL-LarΔF) strain.
Three exconjugants for each strain were chosen for fermentation and LAR analysis. These exconjugants were grown in manually designed antibiotic-screening medium (ASM; composition (g/L); starch (10), casein enzyme hydrolysate (10), ammonium sulfate (10), MgSO4.7H2O (1), NaCl (5), CaCO3 (0.5), KH2PO4 (0.7), K2HPO4 (0.9)) for four days. Crude extracts were prepared using Diaion HP20 resin and subjected to cation-exchange chromatography as described above. The fractions were analyzed by RP-HPLC, and the chromatograms were compared to LAR purified from Paenibacillus sp. M2, with peak identity confirmed by mass spectrometry.
MIC determination
MICs were determined using broth microdilution method in cation-adjusted Mueller-Hinton Broth (MHBII, BD Difco) following standard procedures unless stated otherwise. Mycobacteria susceptibility was tested in Middlebrook 7H9 medium supplemented with 10% OADC Enrichment (oleic acid, bovine albumin, dextrose, and catalase) (BD Difco). The compounds (2 to 4 μl) were mixed with diluted cultures (5–7 x 105 cfu/mL) in MHB (196 to 198 μL) in 96-well round-bottom plates, and 2-fold serial dilutions were made. Plates were incubated at 37°C for 18-22 h, and OD600 readings were taken on Synergy Microplate Reader (Biotek). MIC was determined to be the lowest concentration of antibiotics with no visible growth. For Mycobacterial cultures, the plates were read after two to three days. Susceptibility was also assessed in MOPS minimal medium with 0.4% glucose (M2106 Teknova) and RPMI-1640 medium. Serum effects were tested in MHB with 10% or 50% fetal bovine serum (Invitrogen) or human serum (Heat Inactivated, from human male AB plasma; Sigma-Aldrich). For anaerobic conditions, E. coli was incubated in BD GasPak™ EZ pouch systems. To adjust the pH of the media, 1M NaOH or 1N HCl was used.
For MIC testing using human microbiota strains, the cultures were grown in BHI supplemented with L-cysteine (0.5g/L), Hemin (10mg/L), and Vitamin K (1mg/L) in anaerobic chambers (37 °C, 5% H2, 10% CO2, 85% N2). Candida albicans was cultivated in yeast nitrogen base (YNB) medium without amino acids (BD Difco).
To determine the cross-resistance of LAR with other translation inhibitors, MIC was measured in E. coli BW25113 ΔtolCΔbamB strain expressing various resistance genes as described elsewhere43.
Cytotoxicity of LAR against human cells
HEK293 cells were seeded at 7500 cells/well in 384-well plates with DMEM supplemented with 10% FBS, 2 mM L-glutamine, 100 U/mL penicillin, and 100 μg/mL streptomycin, incubated for 18 h at 37°C in the atmosphere of 5% CO2. DMSO-dissolved compounds or DMSO were added using a Labcyte Echo acoustic dispenser (Beckman Coulter) and a combi nL (ThermoFisher) dispenser, and cells were incubated for 48 h. Cell viability was assessed using Promega Cell Titer Glo 2.0 reagent, with luminescence read on a Neo2 plate reader (Biotek). Wells with no solvent added, or those supplemented only with DMSO were used as controls. A prestoBlue cell viability assay was also used to evaluate cell survival and assess cytotoxicity.
Hemolysis
Human blood (BioIVT, USA) was centrifuged to remove plasma, and erythrocytes were washed with 0.85% NaCl and resuspended in PBS (pH 7.4) to maintain hematocrit. Compounds (1 μl) were added to a 96-well V-bottom plate using Labcyte Echo, with 1% DMSO (final concentration) as a control. Triton X-100 served as a positive control. Erythrocytes were diluted 1:50 in PBS, 99 μl added to wells, incubated at 37°C for one hour, centrifuged at 1000 xg for 5 min, and the optical absorbance of the supernatant was measured at 540 nm on a Neo2 plate reader (Biotek). The release of hemoglobin with Triton-X was considered 100% hemolysis.
Time-dependent killing and cell lysis assay
E. coli BW25113 was grown overnight in 5 ml MOPS minimal medium and diluted 1:100 in 3 ml of fresh medium in 14 ml culture tubes. Cells were grown for three to four hours to reach mid-exponential phase (OD600 0.4-0.6) and treated with 10xMIC of LAR (40 μg/ml) or colistin (2.5 to 5 μg/ml) in 1 ml culture at 37°C with agitation at 250 rpm. Cultures were sampled at various times, centrifuged at 10,000 xg for 3 min, and plated on MHA plates. The number of colony forming units (cfu) was determined after incubating plates for 20-24 hr at 37°C. A time-kill assay against A. baumannii C0286 was performed in MHB. For cell lysis assay, cultures were prepared as described above. Initial OD600 was adjusted to 0.15–0.2, and 98 μl of culture was added to a 96-well plate containing 2 μl of compound. The plates were incubated with orbital shaking at maximum speed with a Synergy Microplate Reader (Biotek), and OD600 was measured over 6 h. Experiments were performed in two to three biological replicates. For colistin in cell lysis assay, clumping of cells was observed after ≈2h, leading to a false increase in OD; therefore, data for colistin was not plotted after this period. The plate was manually mixed on a vortex, and the turbidity of colistin-treated wells was confirmed.
Propidium iodide uptake assay
E. coli BW25113 cells were prepared as described above and mixed with propidium iodide (final concentration 4 μM). Cell suspension (190 μL) was then placed into wells of a 96-well black-wall plate and supplemented with 10 μL of compounds at various concentrations. Fluorescence (lex/lem: 535/617 nm) was monitored for 30 min at RT in a Synergy Microplate Reader (Biotek). Colistin was used as a positive control.
b-galactosidase activity assay
E. coli TOP10 cells with pUC19 plasmid (containing the lacZ gene) were grown to mid-log phase, OD600 adjusted to 0.1, and treated with Ortho-nitrophenyl-β-D-galactopyranoside (ONPG) (1.5 mM). Cells were mixed with compounds in a 96-well plate (final volume 200 μl), incubated at 37°C, and absorbance at 420 nm measured for 30 minutes. Colistin was used as a control.
Membrane depolarization assay
Mid-exponential phase E. coli BW25113 culture (OD600 0.3–0.5) was mixed with 3,3′-Dihexyloxacarbocyanine iodide (DiOC2(3), ThermoFisher Scientific) dye at a final concentration of 30 μM and 190 µL of cells were placed into wells of a 96-well black plate. A volume of 10 μl of test compounds or DMSO was added, mixed by pipetting, and fluorescence was measured for 60 min at room temperature using a Synergy Microplate Reader (Biotek) (lex/lem: 450/670 nm). Carbonyl cyanide m-chlorophenyl hydrazone (CCCP) served as a positive control.
Scanning electron microscopy
Approximately 108 cells of E. coli BW25113 were treated with LAR (40 μg/ml, 10xMIC) in MOPS minimal medium for 1 h at 37°C, centrifuged at 5,000 xg for 5 min and resuspended in 0.1x volume of fixative solution (4% glutaraldehyde in PBS, pH 7.4). Cells were fixed at room temperature for 1 h and stored at 4°C overnight. The next day, 50 μL of the fixed cells were placed on poly-L-lysine coated coverslips, dehydrated through graded ethanol steps, and dried using a critical point dryer. Samples were analyzed using a scanning electron microscope (TESCAN VEGA-II LSU) equipped with an X-MAX 80mm2 EDS detector, and images were acquired using INCA software.
Selection of spontaneous resistant mutants
Approximately 109 cfu of E. coli BW25113 or B. subtilis 168 were plated on MOPS minimal agar and Mueller-Hinton agar, respectively, containing varying concentrations of LAR. Plates were incubated at 37°C for 24-48 h. Colonies that appeared were tested for LAR susceptibility using MIC assays. Genomic DNA from representative resistant mutants (n=3 for E. coli and n=6 for B. subtilis) was isolated and sequenced using Illumina technology. The resulting sequences were compared to the reference genome of the parental strains to identify mutations using breseq44.
To identify LAR-resistance mutations in rRNA genes, E. coli SQ110 ΔtolC (SQ110DTC)45 harboring a single rrn operon was grown overnight in MHB supplemented with 50 mg/mL of kanamycin. Cells were diluted 100-fold into fresh MHB grown until cell density reached OD600 of 0.6. Then, 1 OD600 unit of cell culture (~0.85 x 109 cells) was plated on an MHB/agar plate containing 50 µg/mL kanamycin and 64 μg/mL LAR (~4xMIC). Several dozen colonies appeared after 48 h incubation at 37°C. For 20 randomly selected colonies, rDNA was PCR-amplified using the primers rrnE_F and rrnE_R (Supplementary Table 1) and sequenced. The LAR MIC in liquid MHB medium was then determined for the isolates harbouring mutations in the rDNA (n=8).
Synthesis of LAR-fluorophore conjugates and purification of LAR-B
To protect the free amines in LAR, di-tert-butyl decarbonate (Boc anhydride) was selected as the protecting group. LAR (30 mg, 0.016 mmol, 1 eq) was dissolved in 16 ml of 50% acetonitrile/water. After addition of Boc-anhydride (35 mg, 0.16 mmol, 10 eq) and triethyl amine (TEA, 32.3 mg, 45 µl, 0.32 mmol, 20 eq), the reaction mixture was stirred at room temperature (RT) for 1 h. The reaction was confirmed by LC-ESI-HRMS for the presence of Boc groups on LAR. Two major products were identified as 1186.29 [M+2H]2+ and 1098.74 [M+2H]2+, corresponding to penta-Boc-protected LAR (Boc(5)-LAR) and tetra-Boc-protected LAR-B (Boc(4)-LAR-B) respectively. A small peak corresponding to Boc(5)-LAR-C was also detected. The crude mixture was lyophilized to yield compound 2 (Boc-LAR mix).
To attach a click-chemistry handle on the free carboxyl group of the C-terminus, propargyl amine was selected and amidated as follows: Compound 2 (30 mg, 0.013mmol) was dissolved in 1 mL 50% DMSO/DMF, and to this solution were added propargyl amine (3.6 mg, 0.065mmol, 5eq), TEA (13.1 mg, 0.13 mmol, 10 eq) and PyBop (33.8 mg, 0.065 mmol, 5 eq) as 1 M solutions in 50% DMSO/DMF. The reaction was stirred at RT for 30 min, analyzed by mass spectrometry, and then purified using reversed-phase chromatography (C18, 50 g) on a CombiFlash system (Teledyne Inc.). Water/acetonitrile containing 0.07% TFA was used as the mobile phase. Boc(4)-LAR-B did not react with propargyl amine in the above reaction, indicating that C-terminal carboxyl is not free. The peak corresponding to this compound was resolved from Boc(5)-LAR and Boc(5)-LAR-C on a C18 column (owing to the differences in their hydrophobicity), collected separately and processed as described below to yield pure LAR-B. The other two compounds reacted with propargyl amine successfully and eluted as a single peak that was collected and lyophilized to yield compound 3 (Boc-LAR-alkyne). Boc-deprotection of compound 3 was performed in 5 ml of 20% TFA in DCM at RT for 20 min. The solvent was evaporated under nitrogen gas, resuspended in 50% acetonitrile/water, and lyophilized to obtain compound 4 (LAR-alkyne). Boc(4)-LAR-B was deprotected similarly to yield pure Lar-B. The homogeneity of LAR-B was confirmed by LC-MS (Supplementary Fig. 11a).
For conjugation of the BODIPY or rhodamine dyes, click-chemistry46 was employed as follows: compound 4 (2 mg, 0.001mmol) was dissolved in 0.45 mL of H2O, after which copper(II) sulfate and sodium L-ascorbate were added to the final concentrations of 250 µM and 5 mM, respectively. BODIPY FL azide (Lumiprobe Corporation) or rhodamine azide46 (65 µl from 10 mM stock solution in DMSO) were then introduced into the reaction and allowed to proceed for 30 min at RT with stirring. The reaction was monitored by mass spectrometry, and the final product was purified using RP-HPLC to obtain LAR-BODIPY or LAR-rhodamine. The fluorophore-conjugates were lyophilized and dissolved in DMSO for further experiments.
Fluorescence microscopy
E. coli BW25113 was grown overnight in MOPS minimal medium, then subcultured in fresh medium and grown to mid-exponential phase (OD600 0.3–0.4)). Two µL of LAR-BODIPY or LAR-rhodamine (final concentration 20 μg/mL) from a stock solution in DMSO were added to 0.5 mL of cell cultures and incubated for 1-2 h at 37°C. Twenty min before harvesting the samples, Hoechst 33342 dye was added to the final concentration of 20 μg/mL, and 2 min before harvest, FM-4-64 (final concentration 20 μM) was added. Cells were centrifuged to remove excess dye and the compound and resuspended in 25-50 μL of PBS. Five μL of cell suspension were spotted on PBS agarose pads (1% agarose) and covered with high-precision cover glass (1.5H Thickness, cat. no. NC1415511, Fischer Scientific). The slides were visualized using a ZEISS LSM980 confocal microscope (Zeiss), and the images were acquired and processed using ZEN Blue software.
To test the effect of pH on localization, the pH of the MOPS minimal medium was adjusted using 1M NaOH or 1N HCl, and the cells were subcultured in a modified medium until mid-exponential phase before treatment as described above. For the CCCP assay, LAR-BODIPY and CCCP were added simultaneously. All confocal microscopy experiments were conducted minimally in duplicate.
In vitro transcription/translation assay
The effect of LAR on in vitro protein synthesis was assessed using an E. coli S30 extract transcription-translation system for circular DNA (Promega), with pBESTluc plasmid DNA serving as the template for the firefly luciferase production. Following the manufacturer's protocol, the reactions were carried out in 25 μl in a 96-well plate. LAR was tested at concentrations ranging from 0 to 50 µM. Reactions were incubated for one hour at 37°C. Luminescence was measured in a 96-well opaque plate using Synergy Microplate Reader (Biotek). IC50 values were calculated using GraphPad Prism 10 software.
To test the effect of LAR specifically on translation, sfGFP mRNA was generated by in vitro transcription using MEGAscript™ T7 Transcription Kit (Thermo Scientific) and purified by lithium chloride (LiCl) precipitation method as described in the manufacturer’s manual. In vitro translation was carried out in the system as mentioned above with real-time monitoring of sfGFP fluorescence (lex/lem: 483nm/513nm) for one hour. 1μl of mRNA (from 1μg/μl stock) was added in a 25 μl reaction.
In vitro translation in the rabbit reticulocyte lysate
The effect of LAR on eukaryotic translation was assessed using Rabbit Reticulocyte Lysate System (L4960, Promega) programmed with firefly luciferase mRNA. In vitro reactions were assembled according to the manufacturer’s protocol in a final volume of 10 μL supplemented with 0.02–50 μM of LAR or no antibiotic and incubated for 90 min at 30°C. After that, 2.5 μL from each reaction was mixed with 50 μl of prewarmed luciferase assay reagent, and the luminescence was measured in Infinite M200 PRO plate reader (TECAN) for 10 min at 25°C. The assay was performed in triplicate.
Toeprinting assay
Toe-printing analysis was carried out in the E. coli in vitro transcription-translation system assembled from the purified components (PURExpress, NEB) as described previously47. Reactions either contained no antibiotic or were supplemented with 50 μM retapamulin or varying concentrations of LAR (1, 5, or 10 μM). The model RST1 template encoding a 21-aa long peptide containing all 20 proteinogenic amino acids22 was generated by PCR using the primers RST1_F and RST1_R (Supplementary Table 1).
Translocation inhibition assay
In vitro translocation assay was carried out with a model mRNA with the sequence 5’- AUUAAUACGACUCACUAUAGGGCAACCUAAAACUUACACACGCCCCGGUAAGGAAAUAAAA-AUG-UUC-AAA-GCA-UUC-AAA-AAC-AUC-AUA-CGU-ACU-CGU-ACU-CUU-UAAGCGCAGGCAAGGUUAAUAAGCAAAAUUCAUUAUAACC - 3' encoding the MFKAFKNIIRTRTL peptide (underlined part). The mRNA was prepared by in vitro transcription of a PCR product amplified using the primers MF_F1, MF_F2, and MF_R (Supplementary Table 1). In vitro transcription was performed using HiScribe® T7 High Yield RNA Synthesis Kit (New England Biolabs) as recommended by the manufacturer. A 4.5 μL reaction containing 1 μM E. coli ribosomes, 0.5 μM mRNA, 1 μM tRNAiMet, 0.5 μM radiolabelled NV1 primer (Supplementary Table 1), 2 U/μL RiboLock RNase Inhibitor (Thermo), and antibiotic tested (62.5 μM LAR or 250 μM negamycin) in Pure System Buffer (PSB; 9 mM Mg(CH3COO)2, 5 mM K3PO4, 95 mM potassium glutamate, 5 mM NH4Cl, 0.5 mM CaCl2, 1 mM spermidine, 8 mM putrescine, 1 mM dithiothreitol, pH 7.3)48 was incubated for 20 min at 37°C. Then N-acetyl-Phe-tRNAPhe was added to the final concentration of 2 μM followed by 10 min incubation at 37°C. After that, E. coli EF-G and GTP were added to the final concentrations of 0.2 μM and 533 μM, respectively. After 5 min incubation at 30°C, 1 µL of the mixture of AMV reverse transcriptase (Roche) and dNTPs (2.1 U/µL AMV RT and 2 mM dNTPs in PSB) was added, and the reactions were incubated for another 5 min at 30°C. To stop the reaction, 200 μL of the resuspension buffer (300 mM NaCH3COO, 5 mM EDTA, 0.5% SDS) were added. DNA was then extracted following the phenol-chloroform extraction procedure and precipitated by adding 3 volumes of ice-cold ethanol, incubating at -70°C for 15 min, and centrifugation for 30 min (4°C, 20000 g). The reaction products were resolved in 6% sequencing polyacrylamide gel and imaged on the Typhoon phosphorimager.
Miscoding assay
E. coli strain CA24430 harboring a premature stop codon in the lacZ gene (C1456T), was used in the in vivo miscoding assay. The production of full-length functional β-galactosidase was monitored by the appearance of the blue halo on the indicator agar plates containing X-gal (5-bromo-4-chloro-3-indolyl-β-D-galacto-pyranoside). For the X-gal assay, cells were plated on MOPS agar (Teknova) supplemented with 80 µg/ml X-gal and 250 µM IPTG. Drops of LAR, streptomycin, gentamicin, and chloramphenicol solutions (containing 60.7, 4, 3.1, and 0.9 µg of the antibiotic, respectively) were spotted on the plates. Plates were incubated for ~18 h at 37°C and photographed.
X-ray crystallographic structure determination
Wild-type 70S ribosomes from T. thermophilus (strain HB8) were prepared as described previously49-52. Synthetic mRNA with the sequence 5’-GGC-AAG-GAG-GUA-AAA-AUG-UUC-UAA-3’ containing Shine-Dalgarno sequence (underlined) followed by methionine (AUG) and phenylalanine (UUC) codons were obtained from Integrated DNA Technologies (USA). Non-hydrolyzable aminoacylated Phe-tRNAPhe and fMet-tRNAiMet were prepared as described previously51,53-55.
LAR and LAR-B compounds were soaked into the pre-formed crystals of 70S ribosome in a functional state corresponding to the PTC pre-peptide bond formation with unreacted aminoacylated Phe-tRNAPhe and fMet-tRNAiMet in the A and P sites, respectively. Complexes of the wild-type T. thermophilus tightly-coupled 70S ribosomes with mRNA, aminoacylated A-site Phe-tRNAPhe, aminoacylated P-site fMet-tRNAiMet were formed by programming 5 μM 70S ribosomes with 10 μM mRNA and 20 μM P- and A-site tRNAs. The LAR compound was also soaked into the pre-formed crystals of Tth 70S ribosomes complexed with E. coli protein Y (PY) essentially as described previously55,56. All complexes were formed in buffer containing 5 mM HEPES-KOH (pH 7.6), 50 mM KCl, 10 mM NH4Cl, and 10 mM Mg(CH3COO)2, and then crystallized in the buffer containing 100 mM Tris-HCl (pH 7.6), 2.9% (v/v) PEG-20K, 9-10% (v/v) MPD, 175 mM arginine, 0.5 mM β-mercaptoethanol. Crystals were grown by the vapor diffusion method in sitting drops at 19°C, stabilized and cryo-protected stepwise using a series of buffers with increasing MPD concentrations (25%, 30%, 35%) until reaching the final concentration of 40% (v/v) MPD as described previously 51-53,55,57. Antibiotics LAR or LAR-B were included in all stabilization buffers (250 µM). After stabilization and cryo-protection, crystals were flash-frozen using a nitrogen cryo-stream at 80°K (Oxford Cryosystems, UK).
Collection and processing of the X-ray diffraction data, model building, and structure refinement were performed as described in our recent reports53-57. Diffraction data were collected at beamlines 24ID-C and 24ID-E at the Advanced Photon Source (Argonne National Laboratory). A complete dataset for each complex was collected using 0.979 Å irradiation at 100 K from multiple regions of the same crystal, using 0.3-degree oscillations. Raw data were integrated and scaled using XDS software (version from Jan 10, 2022)58. Molecular replacement was performed using PHASER from the CCP4 program suite (version 7.0)59. The search model was generated from the previously published structures of T. thermophilus 70S ribosome with bound mRNA and aminoacylated tRNAs (PDB entries 6XHW55 or 6XHV55. Initial molecular replacement solutions were refined by rigid-body refinement with the ribosome split into multiple domains, followed by positional and individual B-factor refinement using PHENIX software (version 1.17)60. Non-crystallographic symmetry restraints were applied to four parts of the 30S ribosomal subunit (head, body, spur, and helix 44) and four parts of the 50S subunit (body, L1-stalk, L10-stalk, and C-terminus of the L9 protein). Structural models were built in Coot (version 0.8.2)61. The statistics of data collection and refinement are compiled in Supplementary Table 3. All figures showing atomic models were rendered using PyMol software (www.pymol.org).
Ex vivo blood infection model
A. baumannii C0286 was cultured overnight in MHB and diluted 1:200 in 0.5 ml human blood in 14 ml culture tubes (BioIVT, USA). Five microliters of compound or water were added, and the tubes were treated for 4 hours at 37°C, 220-250 rpm. The remaining cfu were enumerated by appropriate dilution in PBS and spotting on MHA plates. The experiment was conducted in three biological replicates.
Animal studies
All animal studies were conducted according to guidelines set by the Canadian Council on Animal Care using protocols approved by the Animal Review Ethics Board at McMaster University under Animal Use Protocol #20-12-41. All animal studies were performed with 6-10-week-old female CD-1 mice (Harlan/Envigo, Cat. No. 030). Mice were rendered neutropenic by intraperitoneal administration of cyclophosphamide (Sigma-Aldrich, cat.#C0768-5G) at 150 mg kg-1 (body weight) four days before infection, followed by 100 mg kg-1 one day before infection. On the day of infection, mice were infected intramuscularly with 1 109 cfu A. baumannii C0286per thigh, and their core body temperature and body weights were monitored. The treatment group was intraperitoneally administered lariocidin suspended in Ultrapure Distilled Water (Fisher Scientific, Cat. No.10-977-015) at 50 mg kg-1 at 1 h, 4 h, 8 h, and 20 h post-infection. The control group was intraperitoneally administered 10 μL/g (body weight) of Ultrapure distilled water. To quantify bacterial load, mice were sacrificed at 24 h or 48 h, or sooner if they reached the humane endpoint defined by our protocols. Spleen and thigh tissue were harvested and placed in Mixer Mill safeseal tubes containing 1mL of sterile PBS with beads and homogenized using a Mixer Mill at 30 Hz. Blood was collected in Lithium-Heparin Microtainer Tubes (VWR, Cat. No. CABD365965). Bacterial burden was enumerated by plating the tissue homogenates and blood on LB agar plates supplemented with ampicillin (200 μg/mL) and incubating overnight at 37 deg. C.
Statistical methods
Unless stated otherwise in the legend, a two-tailed Mann–Whitney test was used to compare the two groups. A one-way ordinary ANOVA or two-way mixed-effects ANOVA with a Geiser-Greenhouse correction was used for multiple comparisons, as mentioned accordingly. Values were considered statistically significant at p < 0.05. Data were collected using Microsoft Excel, and statistical analyses were performed using Prism GraphPad v. 10.2.3.