Bacterial strains and growth conditions All strains and plasmids utilized in this study are enumerated in Supplementary Table S1, while primers are in Supplementary Table S2. Pseudomonas strains used in this study includes P. aeruginosa PAO1, PA14, P5 and PDO300. Routine cultures for Pseudomonas strains were performed at 37℃ in Luria-Bertani (LB) broth or on LB agar plates with antibiotic where necessary: 20 μg/mL gentamicin and 30 μg/mL apramycin. For gallium treatment CRISPRi-seq screen, PA14 were cultured in M9 minimal medium containing Na2HPO4·7H2O (47.8 mM), KH2PO4 (22 mM), NaCl (8.6 mM), NH4Cl (18.6 mM), MgSO4 (1.0 mM), CaCl2 (0.1 mM), and sodium citrate (20 mM) as the carbon source at 37°C under vigorous aeration. E. coli WM3064 represents a diaminopimelic acid (DAP)-auxotrophic strain (Wang et al. 2015), offering significant utility for conjugation experiments and replication of plasmids necessitating Pir protein. E. coli strains were cultivated in LB broth at 37°C, supplemented with antibiotics where necessary: 20 μg/mL gentamicin and 100 μg/mL ampicillin. Cultures were subjected to CRISPRi repression, primarily induced with 25 ng/mL doxycycline (Dox), unless stated differently.
Development of a tetracycline (tet)-inducible platform in P. aeruginosa
The tet-inducible system for P. aeruginosa was developed based on our previously published tet-inducible system in Streptococcus pneumoniae, which was proved to be very tightly controlled (Sorg et al. 2020; Liu et al. 2021). A gblock (gblock 6-TetPt5) containing Pseudomonas codon-optimized tetR (termed tetR-opt), a constitutive Pcon promoter and tet-inducible Ptet promoter were synthesized by Sangon (Sangon, China). The sequence of gblock 6-TetPt5 is shown in Supplementary Table S3. The tet-inducible system uses a modified pUC18T-mini-Tn7T-P3-luxABCDE-based integration vector (Supplementary Table S1), incorporating a constitutively expressed tetR-opt driven by the Pcon promoter, amplified with primers OXL384 and OXL567 (Supplementary Table S2). The P3 promoter was substituted with a Ptet (amplified with OXL387 and OXL568) to drive luxABCDE reporter gene, enabling precise quantification of tet-regulated gene expression. Herein, we designated the plasmid containing the tet-inducible system as Ptet-lux-Tn7T-Gm. All PCR fragments were amplified using Phanta DNA Polymerase (Vazyme, P515-03, China) in preparation for In-Fusion cloning with the ClonExpress Ultra One Step Cloning Kit V2 (Vazyme, C116-02, China).
Construction of the ccdB counterselection system for sgRNA cloning The ccdB counterselection system (Bernard 1996) was employed to enhance the efficiency of sgRNA cloning through negative selection against non-modified plasmids. Firstly, E. coli WM3064 with mutated gyrA (462Arg→Cys) was engineered via CRISPR/Cas9 editing as previously reported (Chen et al. 2018), to confer resistance to the CcdB toxin (Bernard and Couturier 1992). Sequences of the sgRNA and single strand DNA repair template designed to introduce the gyrA462Arg→Cys mutation by CRISPR/Cas9 editing were listed in the oligo list (Supplementary Table S2). The obtained E. coli WM3064 gyrA462Arg→Cys was confirmed by Sanger sequencing, used as the host for construction of sgRNA cloning vector with ccdB counter selection system. The ccdB gene flanked by two BsaI sites, ordered as the gBlock “gblock4-ccdB-CSY” (Supplementary Table S3) was amplified using primers OXL289 and OXL290, and then cloned into the Mobile-CRISPRi plasmid pJMP2846 (Addgene#160676) (Banta et al. 2020) via BsaI-mediated Golden Gate Assembly. The product of Golden Gate Assembly was transformed into E. coli WM3064 gyrA462Arg→Cys followed by selection with 30 μg/mL apramycin on LB agar plates supplied with 300 μM DAP. The resulting plasmid is named pJMP2846-ccdB, which serves as the vector for constructing the CRISPRi system and subsequent sgRNA cloning.
Construction of the tet-inducible CRISPRi system Our CRISPRi system was primarily constructed based on pJMP2846-ccdB. The tet-inducible element mentioned above was amplified and introduced to replace the native IPTG-inducible part (PLlacO1-LacI) that drives dCas9 expression in pJMP2846-ccdB, using Primers OXL657, OXL658, OXL659 and OXL660 (Supplementary Table S2). The cassette containing Illumina read 1, P3 promoter, BsaI site-flanked mCherry, sgRNA scaffold, sgRNA terminator, and Illumina read 2 was generated from the plasmid pPEPZ-sgRNAclone (Addgene#141090) (de Bakker et al. 2022) using primers OXL226 and OXL227. The kanamycin resistance gene segment was replaced with one conferring apramycin resistance gene using primers OXL483 and OXL484. The construct was then transformed into E. coli WM3064 and selected with 30 μg/mL apramycin on LB agar plate supplied with 300 μM DAP. The resulting plasmid was named as pCRISPRi-ccdB. To test the efficiency of pCRISPRi-ccdB for sgRNA cloning and counter selection, sgRNA targeting different genes were cloned by BsaI mediated Golden Gate Assembly. Two 24-nt oligonucleotides with 5'-ends of TATA and AAAC, respectively, were synthesized for each guide sequence and annealed in TEN buffer (10 mM Tris, 1 mM EDTA, 100 mM NaCl, pH 8.0) in a thermocycler. The annealing process involved incubation at 95°C for 5 minutes and then slowly cooling them to room temperature to allow for proper pairing. The annealed oligonucleotides and pCRISPRi with ccdB vector were assembled based on the One-step Golden Gate protocol (Chen et al. 2018), using DNA restriction enzyme BsaI (Lablead, F5518S, China) and ligase T4 (Vazyme, C301-01, China). The assembly mixtures were transformed into E. coli WM3064 and plated on LB agar plates containing 30 μg/mL apramycin and 300 μM DAP for selection of successful assembly clones. As a control to test the efficiency of CcdB counter selection, the intact pCRISPRi-ccdB plasmid without sgRNA cloning was transformed into E. coli WM3064 in parallel.
Construction of pooled CRISPRi library in P. aeruginosa PA14
sgRNA design
The sgRNAs were designed by our previously published R script for automatic sgRNA selection (https://github.com/veeninglab/CRISPRi-seq) (de Bakker et al. 2022). All sgRNAs were designed to target the coding region of the gene. For each gene, 2 sgRNAs were designed and assigned into two sets. The best sgRNA hits were assigned as set 1, and the 2nd best hit as set 2. For the genes with less than 2 PAMs in the coding regions, only one sgRNA for set 1 was designed. To make the genome-wide sgRNA pool, 5981 sgRNAs were designed in set 1 sgRNA pool and 5971 sgRNAs were designed in set 2 sgRNA pool. The 20-nt spacer sequence of the designed sgRNAs and their targets were listed in Supplementary Table S4.
sgRNA cloning
The set 1 and set 2 pooled sgRNA oligos were synthesized as one oligo chip by GenScript (Nanjing, China), and the oligo sequences were listed in Supplementary Table S4. Each oligo contains a 20-nt sgRNA spacer sequence flanked with 2 BsaI sites, the set specific sequence for amplification of either set 1 or set 2 sgRNAs, and the universal sequence for amplification of all the sgRNAs in one chip. Set 1 and set 2 sgRNA pools were amplified with primer pairs OXL409/OXL410 and OXL411 /OXL412 (Supplementary Table S2) by PCR, respectively. The amplicons were purified with a Monarch DNA Cleanup kit (NEB, T1030S, USA). The purified amplicons were cloned into the plasmid pCRISPRi-ccdB by BsaI mediated Golden Gate Assembly. Specifically, for each Golden Gate Assembly reaction, 300 ng of the amplicons were mixed with 500 ng of the pCRISPRi-ccdB plasmid in a 200-μL PCR tube, followed by addition of 1 μL of with BsaI (Lablead, F5518S, China), 1 μL of T4 ligase (Vazyme, C301-01, China) and 1 µL of 10× T4 ligation buffer. MiliQ water was added into the PCR tube to bring the final volume to 20 µL. The reaction mixture was incubated in a thermocycler with the following program: 150 rounds of 37℃ for 1.5 min, 16℃ for 3 min, and 1 round of 37℃ for 5 min, 80℃ for 10 min. The product of the Golden Gate Assembly reaction was then transformed into chemical competent E. coli WM3064 and the success transformants were selected on LB agar plates with 30 μg/mL apramycin and 300 μM DAP on LB agar plates. In total 60 of 20-uL Golden Gate Assembly reaction products were performed and transformed. Over 250,000 transformant colonies were harvested for each sgRNA pool, indicating more than 50 times sgRNA library coverage.
Construction the CRISPRi library in P. aeruginosa PA14 via triparental conjugation
The E. coli WM3064 with sgRNA library were served as donor strain and subsequently conjugated into P. aeruginosa PA14 strain with a helper WM3064 strain harboring a plasmid (pJMP1039, Addgene#119239) that expresses TnsABCD transposase. The triparental conjugation was performed as describe previously (Banta et al. 2020). The successful transconjugants with integration of Tn7 cassette into PA14 chromosome were selected on LB agar plates supplemented with 30 μg/mL apramycin at 37°C. In total, the CRISPRi library construction involved collecting around 300,000 colonies for the set 1 sgRNA pool and 570,000 colonies for the set 2 sgRNA pool.
CRISPRi-seq Screen
Sample preparations and sequencing
The gene essentiality screen of P. aeruginosa PA14 was conducted over approximately 21 generations of growth in triplicate, and samples of 7 generations, 14 generations and 21 generations were collected and analyzed by Illumina sequencing. Pooled libraries were cultured in LB broth at 37°C until reaching an optical density of 0.6 at 600 nm. The cultures were then diluted 1:100 into fresh LB broth with or without the inducer Dox (25 ng/mL). Upon reaching an OD600 of 0.6 again, 5 mL of bacteria culture was collected as the samples of 7 generations. Consecutively, the cultures underwent two more 1:100 dilutions into fresh LB broth to allow for 14 and 21 generations of Dox induction followed by bacterial pellet collection. Bacterial pellets harvested at 7, 14 and 21 generations were utilized for genomic DNA isolation using a FastPure Bacteria DNA Isolation Mini Kit (Vazyme, DC103-01, China) according to the manufacturer's protocol. For CRISPRi-seq screen under Ga(NO3)3 treatment, pooled libraries were pre-induced in LB broth with Dox for 14 generations, followed by treating with 200 μM Ga(NO3)3 plus Dox in M9 medium and the culture continued until 21 generations. Guide encompassing region was amplified from genomic DNAs using a one-step PCR process, which incorporated Illumina barcodes N701-N712 and N501-N512 as index 1 and index 2, respectively, yielding 303 bp products (de Bakker et al. 2022). The amplicons were sequenced on an Illumina NovaSeq system by Haplox (Shenzhen, China) following the manufacturer's protocol.
CRISPRi-seq data analysis
The absolute abundance of each sgRNA per condition from the raw paired-end sequencing data of the CRISPRi-seq screen is determined with 2FAST2Q (v2.5.0) (Bravo, Typas, and Veening 2022). Using the default configuration of 2FAST2Q, a nucleotide-based quality filtering step discards all trimmed reads with a Phred score below 30. The depletion or enrichment of sgRNAs was analyzed using DESeq2 in R (version 4.3.1) (Love, Huber, and Anders 2014). In the gene essentiality screen, fitness values were calculated by comparing the CRISPRi induction with Dox to the non-induction condition (+Dox Vs -Dox). Gene essentiality was determined by evaluating the log2(Fold change, FC) values from both set 1 and set 2 screening results over 7, 14, and 21 generations. Essential genes were identified for those with log2FC < −1 and adjusted P-value < 0.05. The results from individual analysis of set 1 and set 2 across 7, 14 and 21 generations were displayed in Supplementary Table S5. In the gene categorization, genes were firstly classified into 3 categories, including “nonessential” “vulnerable” and “invulnerable” according to the standard demonstrated in Extended Fig. 2c. The set with the bigger |log2FC| was chosen to classify each gene's essentiality. Within the vulnerable category, genes were further divided into quick- and slow- responsive, based on whether the 7-generation log2FC was less than -2 or greater than -2, respectively. Similarly, within the invulnerable group, genes were classified as quick responders if the 7-generation log2FC was less than -1, and slow responders if it was more than or equal to -1. For genes identified as nonessential, the screening results from set 1 were used as the definitive reference. The essential gene list was compared with three Tn-seq studies conducted in P. aeruginosa PA14 (Liberati et al. 2006; Skurnik et al. 2013; Poulsen et al. 2019). Core essential genes identified by Poulsen et al. (Poulsen et al. 2019) were highlighted in the list. Operon information was extracted from the study by Wurtzel et al. (Wurtzel et al. 2012). The generated results, including the classification of genes into five categories, comparison with Tn-seq studies, and operon information, are presented in Supplementary Table S6.
For the gallium treatment CRISPRi-seq screen, fitness values were determined by comparing the Ga(NO3)3-treated condition with Dox to the untreated condition [+Ga(NO3)3 Vs -Ga(NO3)3]. sgRNA targets were classified as up-regulated genes if their fitness showed a log2FC > 2 with an adjusted P-value < 0.05, and as down-regulated if the log2FC was less than -2 with the same P-value threshold. The results are provided in Supplementary Table S7. GO enrichment analysis for the up- and down-regulated targets was conducted and is presented in Supplementary Table S8.
Construction of knockdown and knockout strains
The gene knockdown strains were engineered using a method akin to library construction but utilized a singular, targeted sequence instead of a diverse sequence pool. The specific gene knockdowns, along with the corresponding primers and bacterial strains, are presented in the results section or supplementary materials. The genes of interest in P. aeruginosa PAO1 were deleted via CRISPR-Cas9 genome editing with pCasPA/pACRISPR system as previously described (Chen et al. 2018). In brief, the pCasPA plasmid expressing Cas9 was first introduced and selected with 100 μg/mL tetracycline. The pACRISPR plasmid containing sgRNA was then electroporated with homologous repair templates flanking the target gene into PAO1 cells harboring pCasPA. The homologous regions were PCR amplified and then purified through gel extraction. Successful knockout mutants were isolated under 100 μg/mL tetracycline and 200 μg/mL carbenicillin selection and validated by sequencing. To achieve gene complementation, the ptet-lux-Tn7T-Gm plasmid, which facilitates gene expression under a tet-inducible promoter, was utilized. The gene's open reading frame was PCR-amplified from PAO1 genomic DNA using primers with 15 bp extensions and subsequently cloned into the ptet-lux-Tn7T-Gm vector through In-Fusion cloning technology (Vazyme, China). This recombinant plasmid was introduced into the WM3064 strain via transformation and selected using 20 mg/mL gentamicin and 300 μM DAP. Following triparental conjugation, the fragment with gene complementation was incorporated into the genome of the PAO1 knockout strain. Complementary mutants were selected on LB agar containing 20 μg/mL gentamicin and further confirmed by PCR and sequencing.
Growth and luminescence measurements
For each strain examined, working stocks were revived and diluted 1:100 in fresh LB broth with or without varying concentrations of Ga(NO3)3 or Dox, to initiate the cultures. The OD600 was standardized to 0.003 for all strains. A total of 100 µL of the bacterial culture were added into 96-well flat-bottom cell culture plates with 3 replicates (NEST Biotechnology, 701011, China) and employed a Tecan Spark microplate reader (Tecan, Switzerland) at 37°C for periodic monitoring of bacterial growth, with an interval time of 10 minutes. For the detection of luxABCDE bioluminescent signals, we transferred the cultures to 96-well flat clear bottom black microplates (αPLUS, WP96-48CMED, China) and recorded measurements at 10-minute intervals at 37°C using the Tecan Spark.
Swarming motility assays Swarming motility assays were conducted based on established methods with specific modifications (Kollaran et al. 2019). The swarming medium was composed of 0.5% LB agar supplemented with 100 µM CaCl2, 2 mM MgSO4, and 0.4% glucose. Each plate was centrally inoculated with 2 µL of bacterial suspension in LB broth, with the culture's OD600 adjusted to 0.2. Plates were then incubated at 37°C for overnight.
Determination of Minimum Inhibition Concentration of Ga(NO3)3 Minimum inhibitory concentrations (MICs) of Ga(NO3)3 were determined using the broth microdilution assay in 96-well polystyrene plates (NEST Biotechnology, 701011, China) as previously described (Dousa et al. 2024). Bacterial suspensions, adjusted to an OD600 of 0.0015, were inoculated into Mueller-Hinton broth (Oxioid, CM0405, USA) containing Ga(NO3)3 at concentrations ranging from 0 to 2560 μM (Oxoid, CM0405, USA). The plates were incubated at 37 °C for 24 hours, followed by visual inspection and OD600 measurement using Tecan Spark.
Gallium time-killing assay For gallium killing assays, bacterial cultures (OD600 = 0.5 ~ 0.7) were diluted to an OD600 of 0.003 in LB broth and incubated at 37°C, with or without Ga(NO3)3 supplementation. An aliquot was plated to determine the colony-forming units (CFU) at Time 0, prior to the addition of Ga(NO3)3. At specified time points, aliquots were taken and washed with sterile PBS. The cells were then serially diluted and plated on LB agar to quantify the surviving bacteria.
Intracellular gallium and iron profiling The methodology for assessing gallium levels in PAO1 strains, including PAO1ΔfprB and complemented strain, was based on the previously established protocol (Tovar-Garcia et al. 2020). Bacterial cultures (OD600 = 0.003) were grown in 6 mL of LB broth, either with or without 10 µM Ga(NO3)3, for 16 hours, and then washed twice using 50 mM HEPES buffer (pH 7.2). The samples were resuspended in 50 mM HEPES buffer and adjusted to an OD600 of 0.6, followed by centrifugation at 8000 g for 5 minutes to pellet the cells. Lysis was performed using 70% nitric acid at 100°C for 1 hour. Sample volumes were then normalized to 2 mL and determined by inductively coupled plasma mass spectrometry (ICP-MS) with a NexION 300X instrument (PerkinElmer, USA). To detect intracellular iron, bacterial cultures (20 mL, initial OD600 = 0.05) were cultivated with or without Ga(NO3)3 at concentrations of 12.5 and 50 µM, and harvested after 16 hours of incubation. These cultures were then sonicated at 200W for 1 minute. Subsequently, cell lysates were prepared for intracellular iron content analysis, employing a previously established method with slight modifications (Zhong et al. 2023). The Total Iron Colorimetric Assay Kit (E-BC-K772-M, Elabscience, China) was used to measure the total iron content in bacteria, and the Ferrous Iron Colorimetric Assay Kit (E-BC-K773-M, Elabscience, China) was utilized to determine the levels of intracellular ferrous iron (Fe2+) using a microplate reader (Tecan, Switzerland) at 593 nm.
Quantification of intracellular ROS and H2O2 by flow cytometry Carboxy-H2DCFDA and PeroxyOrange-1 (Thermo Fisher Scientific, USA) fluorescent probes were employed for detection of total intracellular reactive oxygen species (ROS) and hydrogen peroxide (H2O2), respectively. Upon oxidation, these probes generate fluorescent signals that can be quantified by flow cytometry, thereby indicating the extent of oxidative stress within cells (Hong et al. 2017). Bacterial cultures (OD600 = 0.5 ~ 0.7) were diluted to an OD600 of 0.01 in LB broth, either in the absence or presence of Ga(NO3)3, and incubated for 4 hours at 37°C. Following this incubation, Carboxy-H2DCFDA (final concentration of 10 μM) or PeroxyOrange-1 (final concentration of 5 μM) was added to the growth medium, and the cultures were further incubated in the dark for 2 hours at 37°C with shaking at 250 rpm. An unstained control sample was included to assess the background fluorescence. For each sample, a total of 100,000 ungated events were recorded for each sample. The NovoCyte Advanteon Flow Cytometer (Agilent, USA) was used to measure carboxy-H2DCFDA fluorescence, while the FACSAria II Flow Cytometer (BD Biosciences, USA) was employed for Peroxy Orange-1 fluorescence detection. Data analysis was conducted utilizing FlowJo software version 10 (BD Biosciences).
NADP+ and NADPH detection Total oxidized and reduced nicotinamide adenine dinucleotide phosphates (NADP+ and NADPH) were quantified using the NADP/NADPH-Glo™ Assay (Promega, G9081, UK) according to the manufacturer's protocol. Bacterial cultures were diluted to an OD600 of 0.01 and treated with or without Ga(NO3)3 for 4 hours at 37°C with shaking at 250 rpm. The bacteria were harvested by centrifugation at 7,000 g for 2 minutes and washed with distilled water at 4°C. To ensure complete cell lysis, an equal volume of 0.2 N NaOH containing 1% DTAB (Sangon, A600431, China) was added. Following the manufacturer’s guidelines, NADP/NADPH-Glo™ Detection Reagent was introduced, and luminescence was measured using a Tecan Spark microplate reader.
Protein purification of FprB The fprB coding sequence was cloned into the pGood_6p vector and transformed into E. coli Rosetta (DE3) cells, allowing expression of a GST-tagged protein. Single colony was picked and cultured overnight in LB with 100 µg/mL ampicillin at 37°C and 220 rpm, then diluted 1:100 into 200 mL of LB broth and grown to an OD600 of 0.8. Protein expression was induced with 1 mM IPTG, and cultures were incubated overnight at 16°C with shaking 220 rpm. Cells were harvested by centrifugation, resuspended in buffer (20 mM Tris-HCl, 150 mM NaCl, pH 8.0), and lysed by sonication. The lysate was centrifuged, and the supernatant was incubated with Glutathione Sepharose 4B (Cytiva, 17075605, USA) beads for 4 hours at 4°C. After washing with buffer (20 mM Tris-HCl, 1 M NaCl, pH 8.0), protein was eluted with buffer (20 mM Tris-HCl, 150 mM NaCl, 20 mM GSSH). The GST tag was removed using P3c protease. FprB protein was further purified by size-exclusion chromatography and stored in buffer (20 mM Tris-HCl, 150 mM NaCl, pH 8.0, 20% glycerol).
FprB enzyme activity assay in vitro FprB enzyme activity was assessed using an NADPH-dependent 2,6-dichlorophenolindophenol (DCPIP, from Biosynth, FD32424, Switzerland) reduction assay as previously described (Martinez-Julvez et al. 2017). Briefly, a reaction mixture was prepared in a clear 96-well microplate, containing 800 nM FprB, 400 μM NADPH, 800 µM DCPIP, 2% DMSO, and 50 mM Tris/HCl buffer (pH 8.0). Absorbance at 620 nm was measured using a Tecan Infinite M100PRO reader at 25°C. Negative control wells contained the reaction mixture without FprB. Each condition was tested in triplicate.
RNA-Sequencing P. aeruginosa strains (PAO1 and ΔfprB) at an OD600 of 0.5 ~ 0.7 were harvested and rinsed twice with nuclease-free water. Total RNA was isolated using a RNAprep Pure Cell/Bacteria Kit (TIANGEN, DP430, China). Transcriptome sequencing was performed on an Illumina NovaSeq 6000 platform (Illumina, USA) by Novogene Technology Co., Ltd. (Beijing, China). Differential expression between the two groups was analyzed using the DESeq2 package. Genes with a Benjamini-Hochberg-adjusted P-value < 0.05 and |log2FC| > 2 were classified as differentially expressed.
Distribution, evolution analysis and sequence comparison of FprB
Distribution analysis
Full chromosome sequences of 981 P. aeruginosa strains were downloaded from the NCBI Genome database (https://www.ncbi.nlm.nih.gov/genome, assessed [03/04/2024]). To detect FprB orthologues, the P. aeruginosa PAO1 FprB (PA4615) and its protein sequence (NP_253305.1) were used as the queries. To detect the FprB in the P. aeruginosa genomes, the sequence of NP_253305.1 was used to align against with the peptide sequences derived from the genome sequences of the P. aeruginosa strains. BLASTP was used for the sequence alignment. The criterion for homology was set as ‘coverage * identity’ >= 0.6.
Gene collinearity analysis
The nucleotide sequence containing the fprB gene and its flanking sequences, that is, 5-kb for the upstream and downstream of the center of fprB gene respectively, was retrieved from the genome of each P. aeruginosa strain. PipMaker was used to make the collinearity analysis between each pair of the sequences (http://pipmaker.bx.psu.edu/pipmaker/) (Schwartz et al. 2000).
Multiple sequence alignment and phylogenetic analysis
CLUSTALW was used for FprB protein sequence alignment (https://www.ebi.ac.uk/jdispatcher/msa/clustalo) (Madeira et al. 2024). The FprB multiple sequence alignment results were used for phylogenetic analysis using MEGA 7.0 with the Neigbor-Joining algorithm (https://www.megasoftware.net/) (Kumar, Stecher, and Tamura 2016).
Biofilm assays Biofilm quantification was evaluated in 96-well microtiter plates following established protocols (Haney, Trimble, and Hancock 2021). P. aeruginosa cultures were grown to an OD600 of 0.6. The cultures were then diluted 1:200 in LB broth. Subsequently, 100 μL of the diluted cultures were added to each well, with triplicates for each condition. Following a 24-hour incubation, the media supernatant was discarded, and the wells were washed with distilled water. Biofilms were then fixed with 95% methanol for 15 minutes and stained with 0.5% crystal violet for 10 minutes. The wells were rinsed again with distilled water, and 75% ethanol was added to dissolve the dye. Absorbance was measured at 590 nm after incubation.
Mouse infections The study protocol of animal experiments was approved by the Institutional Animal Care and Use Committee of Shenzhen University Medical School (Approval Number: IACUC-202400111). Female BALB/c mice (18 ~ 22 g, 6 ~ 8 weeks old) were purchased from Guangdong Medical Laboratory Animal Center and maintained under specific pathogen-free conditions. Prior to infection, mice were randomly assigned to either treatment or control groups. Mice were anesthetized and intranasally inoculated with 107 CFU in 40 µL of cell suspension. After 3 hours, mice were treated intraperitoneally with either 50 µL PBS or 250 mM Ga(NO3)3. At 27 hours post-infection (h.p.i.), mice were necropsied and lungs were collected for bacteriological analysis. Lung tissue samples were homogenized, serially diluted in sterile PBS, and plated onto LB agar to enumerate bacterial load.
Statistical analysis GraphPad Prism (version 8.4.3) and R (version 4.3.1) were used for all statistical analyses. The experiments were conducted in at least triplicate to ensure biological reproducibility. Reported values represent the mean and standard deviation (SD) across the three replicates. Statistical significance between groups was determined by Two-way-measure of variance (ANOVA) and the Turkey's multiple comparisons test at a p-value threshold of 0.05. The illustrative diagrams were created or generated using Adobe Illustrator 2021 or BioRender (https://www.biorender.com/).
Resource availability
Materials availability
The vectors pCRISPRi-ccdB and ptet-lux-Tn7T-Gm were deposited at Addgene (catalog #226432 and 226434). The pooled CRISPRi sgRNA libraries (set 1 and set 2) for P. aeruginosa PA14 will be available through Addgene (#xxxxx, under processing).
Data availability
All sequenced data supporting the findings of this study can be accessed through the Sequence Read Archive database. The data is linked to BioProject no. PRJNA1063516 and can be found using accession numbers SRR27547817 to SRR27547879.