Cell line generation and culture conditions
All SKOV3 cell lines and SKBR3 cells were cultured at 37°C with 5% CO2 in RPMI 1640 medium supplemented with 10% fetal bovine serum (FBS) and Glutamax. Parental SKOV3 and SKBR3 lines were obtained from ATCC. RTK-FP SKOV3 cells (Sigma-Aldrich) were subcloned using FACS to yield a homogenously expressing clonal line.
RTK-FP PQLC2KO cells were generated by infection of RTK-FP cells with lentivirus containing lentiCRISPR v2 encoding Cas9 and sgRNAs targeting exon 3 or exon 2 of the SLC66A1 gene. Virus-containing media from HEK293T cells was added at a 1:1 v/v ratio to RTK-FP cells with 5 µg/mL Polybrene transfection reagent. The medium was changed after 24 hrs. After an additional 24 hrs of incubation, infected cells were selected by treatment with 2 µg/mL puromycin for 72 hrs to generate a knockout pool. Clonal lines expressing sgRNA #1 or sgRNA #2 were isolated by limited dilution cloning and knockout of PQLC2 in individual clones was confirmed using genomic sequencing. SKOV3 PQLC2KO cells were similarly generated by infection of SKOV3 cells with LentiCrisprV2 encoding PQLC2 sgRNA #1, selection with puromycin and subcloning. Control cells were generated by infection of RTK-FP or SKOV3 cells with lentivirus containing LentiCRISPR v2 encoding an sgRNA targeting the cell surface protein CD81.
RTK-FP PQLC2KO cells expressing PQLC2 rescue constructs or LAMP1-SNAP were generated by infection with a dilution series of lentivirus containing a construct encoding PQLC2-Flag or LAMP1-SNAP under control of a dox-inducible promoter. Infected cells were selected by treatment with 10 µg/mL blasticidin for 7 days and the multiplicity of infection (MOI) was determined by CellTiter-Glo assay (Promega). Lines infected with a dilution that yielded an MOI ≤ 0.3 were used for downstream experiments. LAMP1-SNAP-expressing cells were further enriched by FACS. Prior, expression of LAMP1-SNAP was induced by treatment with 500 ng/mL doxycycline for 48 hrs and LAMP1-SNAP was fluorescently labeled by treating cells with 1 µM SNAP-Cell 647-SiR (New England Biolabs) for 30 min at 37°C. Cells were pelleted and washed twice prior to sorting of the 647-SiR-positive population.
PQLC2KO-pH cells were generated by infection of SKOV3 PQLC2KO cells with lentivirus containing a construct encoding mScarlet-LAMP1 under control of a dox-inducible promoter, followed by selection with 10 µg/mL blasticidin. mScarlet-LAMP1-expressing clones were isolated by FACS. PQLC2KO-pH expressing PQLC2 rescue constructs were generated by infection with virus containing a construct encoding PQLC2-SNAP under dox control. PQLC2-SNAP-expressing cells were enriched by labeling cells with SNAP-Cell 647-SiR, as above, followed by FACS.
Plasmids
CRISPR guide RNA (sgRNA) sequences targeting the SLC66A1 gene were obtained from the Avana library. The oligonucleotide sequence preceding the protospacer is 5’ caccgCATCTCTTACAGACCTACA 3’ for sgRNA #1 and 5’ caccgGAGACTCCTGCAACCTCAT 3’ for sgRNA #2. For the sgRNA sequence targeting the CD81 gene, the oligonucleotide sequence preceding the protospacer is 5’ caccg tggcttcctgggctgctacg 3’. Nucleotides in lowercase show the overhangs that allow cloning the oligonucleotides into the BsmBI site of lentiCRISPR v2.
The dox-inducible PQLC2 lentiviral expression construct was generated by inserting PQLC2-myc-flag (OriGene Technologies) between the NheI and NotI sites of the custom dox-inducible lentiviral expression vector pSHUSHV2, encoding a blasticidin resistance cassette and the reverse tetracycline-controlled transactivator. PQLC2 mutants were generated by site directed mutagenesis followed by insertion into pSHUSHV2. PQLC2 constructs containing N and C-terminal Myc or Flag tags were generated by incorporating sequences encoding these tags into the PCR amplification primers. PQLC2 containing a C-terminal SNAP tag was generated by Gibson cloning, followed by insertion into pSHUSHV2. LAMP1-SNAP was generated by amplification of the LAMP1 cDNA sequence (Origene Technologies) and the SNAP DNA sequence (New England Biolabs) followed by joining using Gibson assembly and insertion into pSHUSHV2. mScarlet-LAMP, previously described27, was amplified and inserted into pSHUSHV2.
Western blotting
Cells were harvested by scraping or trypsinization and washed with PBS after pelleting by centrifugation. Cell pellets were lysed in 1% SDS/water and solubilized by sonication. Cell lysates were boiled at 100 ºC for 10 min, and protein concentrations were determined using Bicinchoninic acid (BCA) assay (Thermo Fisher Scientific). 20–40 µg of lysate was combined with Laemmli buffer containing 2.5% v/v ß-Mercaptoethanol (BME), heated at 65C for 10 min and separated by SDS-PAGE on 4–20% Criterion TGX gels (Bio-Rad) at 150V for 75 min. The proteins were transferred to 0.22 µm nitrocellulose membranes (Bio-Rad) using the Trans-Blot Turbo transfer system (Bio-Rad). Membranes were blocked in 5% BSA/PBST for 30 min at room temperature followed by overnight incubation at 4 ºC in primary antibodies diluted in 5% BSA/PBST. The following day, membranes were washed 3 times for 5 minutes in PBST, and incubated in secondary anti-mouse IRDye 680RD or anti-rabbit IRDye 800CW antibodies (LI-COR) for 30 minutes at room temperature. After three washes in PBST, the membranes were imaged on an Odyssey Imaging System (LI-COR).
The following primary antibodies were diluted 1:1000 v/v, unless otherwise noted, in 5% BSA/PBST and used for immunoblots in this study: anti-Her2 (Cell Signaling Technology cat no. 4290S), anti-EGFR (Cell Signaling Technology no. D38B1), anti-GAPDH diluted 1:5000 (Cell Signaling Technology no. D7166S), anti-Phospho-Her2 (Tyr1196) (Cell Signaling Technology no. 6942S), anti-Flag (Sigma-Aldrich no. F1804), anti-Myc-Tag (Cell Signaling Technology no. 2272S), anti-WDR41 (Novus Biologicals no. 83812), anti-ß-actin (Cell Signaling Technologies no. 3700s) and anti-LC3 (MBL International Corporation no. PM036).
Microscopy
Live RTK-FP cells expressing LAMP1-SNAP were imaged using a spinning disk confocal microscope (Nikon) equipped with a 60x 1.4na oil objective, in an environmental chamber set to 37 ºC and 5% CO2. Cells were seeded at a density of 10,000 cells per well in a glass bottom #1.5H-N 96-well plate (Cellvis) containing 100 µL of complete phenol-red free RPMI 1640 per well. 500 ng/mL of dox was added to the media to induce expression of LAMP1-SNAP. After a 48 hr incubation, cells were treated with 1 µM Lap-VHL or Lap-VHLInactive and 100 nM Bafilomycin-A1 (Selleck chemicals) for 24 hrs. To label LAMP1-SNAP immediately prior to imaging, cells were treated with 1 µM SNAP-Cell 647-SiR for 30 min at 37 ºC, and subsequently washed three times with complete media while adhered to the imaging chamber. After an additional 30 min incubation, the media was replaced with fresh media containing 1 µg/mL Hoechst 33342 Trihydrochloride Trihydrate (Invitrogen) to stain nuclei.
Time lapse imaging of live RTK-FP cells was performed using a Opera Phenix Plus System (Revvity Health Sciences) set to 37 ºC and 5% CO2. Cells were seeded as above and treated with 1 µM Lap-VHL in combination with degradation inhibitors. To label nuclei, cells were treated with 50 nM SiR-DNA (Cytoskeleton, Inc.) and single plane images across 20 fields per well were acquired in confocal mode every 30 min. At each time point, the number of puncta in each well was determined using Harmony imaging and analysis software (Revvity) and normalized by the number of SiR-DNA-positive nuclei in the well.
For immunofluorescence microscopy analysis of PQLC2-Flag expression, RTK-FP cells were seeded as above and incubated for 48 hrs in the presence of 500 ng/mL dox. Cells were washed three times with PBS and fixed with 4% paraformaldehyde in PBS for 15 min at room temperature. Following fixation, cells were washed three times with PBS, and permeabilized with 0.1% Triton X-100 in PBS for 5 min at room temperature. Cells were blocked in 1% BSA in PBS for 30 min and stained for 2 hrs with a 1:500 v/v dilution of anti-Flag monoclonal antibody (M2, Sigma-Aldrich) in blocking solution. After 3 washes with blocking solution, cells were stained for 1 hr with anti-Mouse IgG Fragment conjugated with Alexa Fluor 647 (Cell Signaling Technologies) diluted 1:5000 v/v in blocking solution, washed 3 times, and imaged on the Opera Phenix Plus System.
Analysis of SKBR3 cell proliferation was performed using an Incucyte SX5 live-cell imaging and analysis instrument (Sartorius). 5000 cells per well were seeded in a 96-well tissue culture plate and incubated for 24 hrs prior to drug treatment and start of imaging. Images were taken in the phase channel every 6 hrs using the 10X objective over a total period of 120 hrs, and percent confluence over time was determined using the Incucyte image analysis software.
Flow cytometry
Flow cytometric analysis was performed using the LSRFortessa X-20 cell analyzer (BD Biosciences). For analysis of total RTK-FP levels, cells were collected from 96-well plates by trypsinization, transferred to V-bottom plates and run using the high throughput sampler. For staining of surface RTKs, trypsinized cells were transferred to V-bottom plates and pelleted at 300 x g for 5 min. Cells were resuspended in 50 µL of a solution containing 0.5% v/v BSA in PBS, 2 µg/mL anti-EGFR Alexa Fluor 405-conjugated antibody (R&D Systems) and 2 µg/mL anti-Her2 Alexa Fluor 647-conjugated antibody (R&D Systems). After a 30 min incubation at room temperature, 100 µL of 0.5% BSA/PBS was added to each well and the plates were centrifuged at 300 x g for 5 min. Plates were washed once with 150 µL of 0.5% BSA/PBS, and the cells were resuspended in FACS buffer containing 2% FBS in PBS for analysis.
Geometric mean fluorescence intensity (MFI) was calculated using FlowJo analysis software (FlowJo, LLC). Each MFI value was normalized by subtracting the background MFI determined from analysis of parental SKOV3 cells and divided by the value obtained from untreated cells within each treatment condition.
Mass spectrometry
To prepare lysates for immunoprecipitation, two replicate samples of RTK-FP PQLC2KO cells, RTK-FP PQLC2KO cells expressing PQLC2-Flag or PQLC2KO cells expressing PQLC2(2x PL)-Flag were treated with 500 ng/ml dox for 48 hrs. After pelleting, cells were washed in cold PBS and lysed in cold RIPA buffer containing 50 mM Tris-HCl pH 7.5, 150 mM sodium chloride, 1 mM EDTA, 1% Triton X-100, 1% Sodium deoxycholate and 1X Protease inhibitors (Thermo Fisher Scientific). Cells were rotated in lysis buffer for 30 min at 4ºC, and lysates were cleared by centrifugation at 20,000 x g for 15 min. Supernatants were transferred to a new tube, and protein concentrations were quantified using BCA assay.
For each replicate, 10 mg of lysate was combined with 40 µL of anti-Flag M2 magnetic beads (Sigma-Aldrich) slurry and rotated for 2 hrs at 4ºC. Beads were separated from lysates using a magnetic tube rack, washed 5 times with cold RIPA buffer, and 2 times with cold wash buffer containing 150 mM Sodium chloride, 10 mM Tris HCl pH 7.5 and 0.5 mM EDTA. Bound proteins were eluted in 40 µL of a 100 µg/mL solution of 3X Flag peptide (Sigma-Aldrich) resuspended in TBS, by incubation for 15 min at room temperature.
Sample preparation and mass spectrometric analysis was performed by Creative Proteomics (Shirley, New York, USA). Eluted proteins were separated on an 12% SDS-PAGE gel and visualized by silver staining. To prepare proteins for in-gel digestion, each gel slice was cut into 1 mm3 cubes and combined with 1 mL of a solution containing 1:1 v/v ratio of 30 mM Potassium ferricyanide and 100 mM Sodium thiosulfate until destaining was complete. The supernatant was removed, and the gel slices were combined with 200 µL water for 10 min followed by 1 mL of 50 mM Ammonium bicarbonate for 30 min. Gel slices were subsequently dehydrated by incubation in 500 µL of Acetonitrile for 30 min. After removal of Acetonitrile, the gel slices were rehydrated with a solution containing 10 mM Dithiothreitol (DTT), and incubated at 56ºC for 1 hr. The DTT solution was removed, and the gel slices were incubated in 500 µL of Acetonitrile for 10 min at room temperature, followed by incubation in a solution containing 50 mM Iodoacetamide at room temperature in the dark. After removal of Iodoacetamide and incubation in 500 µL of Acetonitrile for 10 min, gel slices were incubated in trypsin digestion solution for 45 min on ice, followed by incubation at 37ºC overnight. The supernatant was transferred to a new tube, and the slices were incubated in a 1:2 v/v 50 mM Ammonium bicarbonate:Acetonitrile solution for 1 hr at 37ºC. This procedure was repeated, and all peptide extractions were combined and acidifed with 0.1% Trifluoroacetic acid. After lyophilization, the digested peptides were resuspended in 20 µL of 0.1% Formic acid for LC-MS/MS analysis. Peptides were
separated on a UltiMate 3000 nano UHPLC system (Thermo Fisher Scientific) using a PepMap C18, 100Å, 100 µm × 2 cm, 5 µm trapping column and a PepMap C18, 100Å, 75 µm × 50 cm, 2 µm analytical column. 1 µg of sample was run at a flow rate of 250 nL/min using a gradient of 2–8% of buffer B (0.1% formic acid in 50% Acetonitrile) for 3 min, 8–20% Buffer B for 56 min, 20–40% buffer B for 37 min, and 40–90% buffer B for 4 min.
Samples were analyzed on a Q Exactive HF mass spectrometer (Thermo Fisher Scientific, USA) equipped with an ESI nanospray source. The full scan was performed between 300-1,650 m/z at the resolution 60,000 at 200 m/z. The automatic gain control target for the full scan was set to 3 x 106. The MS/MS scan was operated in Top 20 mode using the following settings: resolution, 15,000 at 200 m/z; automatic gain control target, 1 x 105; maximum injection time, 19 ms; normalized collision energy, 28%; isolation window, 1.4 Th; charge state exclusion, unassigned, 1, > 6; dynamic exclusion, 30 s.
Six raw MS files were analyzed and searched against the Homo sapiens protein database using Maxquant software (version 1.6.2.6). The parameters were set as follows: the protein modifications were carbamidomethylation (C) (fixed), oxidation (M) (variable); the enzyme specificity was set to trypsin; the maximum missed cleavages were set to 2; the precursor ion mass tolerance was set to 10 ppm; MS/MS tolerance was set to 0.5 Da.
Edman sequencing
RTK-FP PQLC2KO cells expressing PQLC2-Flag were treated with 500 ng/mL dox for 48 hrs, and lysates were prepared as described above. 60 mg of lysate in RIPA buffer was combined with 150 µL of anti-Flag M2 magnetic bead slurry (Sigma-Aldrich) and rotated for 2 hrs at 4ºC. Beads were separated from lysates using a magnetic tube rack, washed 5 times with cold RIPA buffer, and bound proteins were eluted by boiling beads in 2X Laemmli buffer for 10 min. The supernatant was transferred to a new tube, combined with 2.5% v/v ß-Mercaptoethanol, and proteins were separated by SDS-PAGE using a 12.5% Bis-Tris gel (Bio-rad) at 150V for 75 min. The proteins were transferred to 0.22 µm PVDF membranes (Bio-Rad) using the Trans-Blot Turbo Transfer System (Bio-Rad). The membranes were stained with Coommassie Gel Code Blue at room temperature for 10 min, and destained several times with 50% methanol. A PVDF band corresponding to the expected size of PQLC2-Flag was cut from the membrane and further destained with 50% methanol in a tube, followed by two washes with water. 10 cycles of Edman sequencing analysis was performed (Creative Proteomics) using an ABI Procise 494HT protein sequencing system (Thermo Fisher Scientific).
Morphological profiling
Cells were seeded inn384-well PhenoPlates (Perkin Elmer) using a Fluent Liquid Handler (Tecan). Prior to seeding, cells were counted using a Vi-Cell XR cell counter (Perkin Elmer), and their concentration was adjusted to generate the required stock concentrations that varied depending on the final time point of analysis. 50 µL of cell suspension was added to each well at a density of 2500 cells per well for the 2 hr and 6 hr time points, 1500 cells per well for the 24 hour time point, 1000 cells per well for the 48 hour time point, and 500 cells per well for the 72 hour time point. After seeding, the plates were left at room temperature for 30 min to allow uniform attachment of cells to the plate. The plates were subsequently transferred to an incubator for 16 hrs.
Cells were treated with experimental compounds using the Echo 555 Acoustic Liquid Handler (Beckman Coulter). Specifically, low dead volume Echo source plates containing 10 µL of compound per well were prepared and stored at -80°C. Source plates were limited to a maximum of three thaws at room temperature and centrifuged at 1000 rpm for 5 min prior to dosing. After dosing, Phenoplates were returned to an incubator for the duration of the 2–72 hr treatments. The dose order for small molecules corresponds to the following concentrations: 0, 0.05 µM; 1, 0.1 µM; 2, 0.35 µM; 3, 1 µM; 4, 3.5 µM; 5, 10 µM. The dose order for antibodies corresponds to the following concentrations: 0, 0.3 µg/mL; 1, 1 µg/mL; 2, 3.3 µg/mL; 3, 10 µg/mL; 4, 33 µg/mL; 5, 100 µg/mL.
After incubation in drugs, the cells were stained with 0.5 µM of MitoTracker Deep Red FM (Invitrogen) in cell culture media by adding 10 uL of a 3 µM MitoTracker solution to each well using a Multidrop Combi Reagent Dispenser (Thermo Fisher). The cells were then incubated at 37°C for 30 min. During this time, the cell staining solution was prepared as follows: 1% BSA solution was made by dissolving fresh Bovine Serum Albumin (BSA) powder in Hank’s Balanced Salt Solution (HBSS) followed by filtration through a 0.22 µm filter. Next, a 0.1% v/v Triton X-100 solution was prepared by mixing 1% BSA/HBSS with a 10% Triton X-100 stock solution. Dyes from the Cell Painting panel were manually added at the following concentrations to generate the post-fixation permeabilization and staining solution containing 1 µg/mL Phalloidin Conjugates CF430 (Biotium), 1 µg/mL Hoechst 33342 Trihydrochloride Trihydrate (Invitrogen) and 1.5 µg/mL Wheat Germ Agglutinin (WGA) Alexa Fluor 555 (Invitrogen). To stain Her2, Trastuzumab (Herceptin, Genentech) and Pertuzumab (Perjeta, Genentech) were labeled using the DyLight 488 Antibody Labeling Kit (Life Technologies Corporation) and added to the staining solution at a concentration of 1 µg/mL. Lastly, the staining solution was combined with 0.05% v/v sodium azide to prevent contamination during staining. After staining mitochondria, the plates were transferred with lids removed to a BioTek EL406 Microplate Washer Dispenser with BioStack (Agilent). 20 µL of a 16% paraformaldehyde (PFA) solution was added to each well using a syringe pump to yield a final concentration of 4% PFA. Cells were fixed for 20 minutes at room temperature and PFA was removed using a wash manifold on the EL406. Excess PFA was removed by washing each well twice with 60 µL of HBSS. Cells were subsequently incubated in 20 µL of the permeabilization and staining solution for 30 min at room temperature followed by two washes with HBSS. Upon completion of staining, all wells were filled with 60 µL of an HBSS solution containing 0.05% v/v sodium azide, and the plates were sealed before imaging. Each stained 384-well plate was imaged using a Opera Phenix Plus System (Revvity Health Sciences) by acquiring 5-channel fluorescence and digital phase contrast images across 9 fields in the center of each well with a 20X objective.
For generation of deep learning image embedding, images were down sampled to 512 x 512 x 6, with each channel corresponding to an image acquisition channel. Separate deep convolutional neural networks were trained using all images for each cell type. All networks were a modified ResNet35 backbone trained using ArcFace loss36, with each image’s class label corresponding to a combination of its timepoint x dose x perturbation. This training objective encourages the network to minimize differences in representation due to batch effects between technical replicates. For downstream analysis, the 512-dimensional final activation vector of the network was used as the image embedding. All image embeddings were further normalized by subtracting the mean of all DMSO embeddings per plate. For analysis in the absence of Her2 staining, a separate network was trained without including the Her2 channel in the input images.
For cell segmentation and image intensity analysis, all channels other than the Hoechst nuclei channel were grouped as “cyto” channels. Cyto channels were combined by first linearly rescaling each individual channel image to have its 3rd to 97th percentile pixel intensity values fall in the range [0, 1], with all outliers clipped to 0 or 1. The channels were then combined by taking the per pixel maximum value across all rescaled cyto channel images. The nuclei channel and composite cyto channel were then down sampled to 512 x 512 and processed using the Cellpose model37 to derive individual cell segmentation masks. For each channel, the median intensity value per cell was then computed.
t-SNE plots were generated by averaging the image embeddings across the timepoint x dose x perturbation groups for each plate. The two dimensional t-SNE embeddings were generated using a multicore implementation of the Barnes-Hut t-SNE algorithm38,39. The perplexity value was manually tuned for each dataset.
Functional genomic screens
For the EGFR-RFP and Her2-GFP BiDAC screens, RTK-FP Cas9 cells were seeded in 5-layer flasks (Corning) at a density of 11 million cells per flask. After 72 hrs, the cells were infected with the Avana sgRNA library40 at a multiplicity of infection of 0.3 and treated with 5 µg/ml of Polybrene transfection reagent (Fisher Scientific). The media was changed after 24 hrs, and cells were incubated for an additional 24 hrs. 48 hrs after infection, cells were treated with 2 µg/ml of puromycin to initiate selection. 48 hrs after addition of puromycin, the puromycin-containing media was replaced with fresh media lacking puromycin and the cells were allowed to recover for an additional 72 hrs. After recovery and 9 days following infection, cells were treated with 1 µM of Lap-VHL and incubated for 24 hrs in BiDAC. Prior to FACS, 60 x 106 cells were removed to serve as a non-sorted control. For enrichment by FACS, cells were harvested by trypsinization in TrypsinLE and resuspended in FACS buffer composed of PBS containing 2% FBS. The top 1% RTK-FP-expressing cells were sorted using a Bigfoot Cell Sorter (Thermo Fisher Scientific), comprising a total of 750 x 103 cells. After collection, cells were pelleted and frozen at -80°C.
To prepare genomic DNA, sorted cells were mixed with 5 x 106 non-infected RTK-FP cells to increase recovery, and the sorted and unsorted cell pellets were resuspended in 100 µL of TE buffer (10 mM Tris-HCl pH 8.0, 10 mM EDTA) per 5 x 106 cells. 900 µL of Lysis Buffer containing 10 mM Tris-HCl pH 8.0, 10 mM EDTA, 0.5% SDS and 0.2 mg/mL Proteinase K (Thermo Fisher Scientific) was added to 100 µL of cells. Cell lysates were incubated at 56°C and vortexed vigorously every 15 min, followed by an additional overnight incubation. After the incubation, 5M NaCl was added to lysates to yield final concentration of 0.2M NaCl. A phenol chloroform extractions was performed using MaXtract tubes (Qiagen) by mixing lysates with an equal volume of Phenol:Chloroform:Isoamyl alcohol 25:24:1 (Life Technologies), followed by centrifugation at 1500 x g and extraction by an equal volume of chloroform. The aqueous phase was treated with 25 µg/mL of RNAse A (PureLink, Life Technologies) for 1 hr at 37°C with periodic mixing followed by a second phenol chloroform extraction. The aqueous phase was combined 1:30 v/v with 3M Sodium acetate, pH 5.2 and the DNA was precipitated with 2.5 volumes of ethanol for 1 hr at room temperature. Precipitated DNA was pelleted by centrifugation at 4,000 x g for 1 hr at 4°C in a swinging bucket centrifuge. The DNA pellet was washed 3 times with 75% ethanol, allowed to dry and resuspended in 200 µL of water per 5 x 106 lysed cells. DNA was further solubilized by incubation for 1 hr at 55°C followed by passage 10–20 times through a 28 gauge needle. DNA purity was assessed using a NanoDrop spectrophotometer and its concentration was determined using Qubit Fluorometric Quantification (Thermo Fisher Scientific).
Genomic sgRNA libraries were prepared by PCR amplification of all DNA extracted from sorted cells and 70 µg of DNA extracted from the unsorted control samples using common primers. 4 µl of the PCR products was used as a template for another round of PCR with barcoded primers. The products were purified using SPRIselect beads (Beckman Coulter), and their purity and concentration was determined using the 2100 Bioanalyzer instrument (Agilent). The libraries were sequenced at a read depth of 60 x 106 reads per sample using a NovaSeq 6000 System (Illumina). MAGeCK was used to normalize the read counts by the median sgRNA count values, calculate fold changes and determine statistical significance16.
Fluorescence Lifetime Imaging Microscopy (FLIM)
Cells were seeded at a density of 10,000 cells per well in a Cellvis 8-well chambered coverglass and treated with 500 ng/ml dox for 48 hrs. To image SNAP-tagged PQLC2, cells were treated with 1 µM SNAP-Cell 647 SiR for 30 min and washed 3 times with complete RPMI 1640 medium. The medium was replaced again following a 30 min incubation with complete RPMI 1640 lacking phenol red. During imaging, cells were incubated at 36ºC and 5% CO2 using a stage top incubator (Okolab).
For pH calibration experiments, cells were imaged immediately after a 5 min incubation in high potassium imaging buffer containing 140 mM KCl, 1 mM CaCl2, 1 mM MgCl2, 5 mM glucose and 10% Carmody’s buffer41 to set the desired pH. Ionophores consisting of 10 µM nigericin and 2 µM monensin was also added to the imaging buffer to equalize proton gradients across cellular membranes.
Fluorescence lifetime imaging was performed on an SP8 point scanning confocal microscope (Leica) equipped with a time-correlated single photon counting (TCSPC) add-on (PicoQuant GmbH, Berlin, Germany). mScarlet was excited with the 561 nm output of the white light laser at an average power of approximately 10–20 µW and a repetition rate of 40 MHz. Emission was collected with a 100x/1.4 NA oil objective (HC PL APO CS2, Leica), and photon arrival times were determined with the built-in HyD detectors and the PicoHarp 300 TCSPC module. Spectral detection was used to restrict collection wavelengths to 590–700 nm, and a notch filter was included to block 488/561/633 nm. Scanning was performed with the built-in Leica non-resonant scanner with a total time per field of view of 20 s. Images were binned to pixel sizes of 200 nm x 200 nm to obtain adequate photons for lifetime determination. All imaging was performed with the confocal pinhole set to 1 Airy unit at 580 nm.
For two color imaging with SiR-SNAP647, we took two sequential images of each field of view. We imaged mScarlet with the 561 nm output of the white light laser collection from 587 to 625 nm. We imaged SiR647 SNAP with the 640 nm output of the white light laser (average power 0.5 µW) with collection from 660 to 788 nm. An additional notch filter (470/640 nm) was included to block the 640 nm illumination. To reduce motion artifacts from lysosomal movement, we alternated between the two illumination colors between lines. Because the PicoQuant software did not support between line switching, we used a custom parser to process the raw TCSPC output and reconstruct an image. Parser code is available on GitHub (https://github.com/AndrewGYork/tools/blob/master/picoquant_tttr.py). For two color images, the total acquisition time per field of view was approximately 40 seconds.
TCSPC images were used to calculate mean arrival time at each pixel either within the PicoQuant SymPhoTime software (one color imaging) or with custom parser code (two color imaging). Fluorescence intensity images were generated by summing the photons at each pixel over the fluorescence lifetime decay.
To segment fluorescence lifetimes and intensity images, we first performed instance segmentation of lysosomes using the mScarlet-LAMP1 emission channel and the “StarDist2D” model trained on the “2D_versatile_fluo” dataset42. The model distinguishes individual lysosomes within 2D images by recognizing their star-convex shape. Prior to segmentation, images were preprocessed by a normalization to adjust the pixel intensity distribution to a standard range and by up sampling to adjust image features to the scale of those used in the training model. After segmentation, the masks were down sampled back to the original size. The segmentation output was used to extract lysosome area as the total number of pixels in the masks scaled by the camera pixel resolution, mean fluorescence values in both the mScarlet-LAMP1 and PQLC2-SNAP channels, and the mean fluorescence lifetime in the mScarlet-LAMP1 channel. These quantifications were performed using Scikit Image’s region properties function43. For fluorescence lifetime quantification, only those pixels with a photon count above 5 were used to calculate the mean florescence lifetime value.
pH calibration (ionophore) data were fit to a four parameter logistic function to extract a calibration curve, where tau is the lifetime, min and max are the minimum and maximum lifetime values observed (at low and high pH respectively), and b is the slope factor (similar to the Hill coefficient).
$$tau=\text{max}+ \frac{\text{min}- max}{1+ {\left(\frac{pH}{pKa}\right)}^{b}}$$
Median lifetime of all lysosomes in each condition was converted to pH using the corresponding calibration curves. For the PQLC2-SNAP rescue experiments, the PQLC2KO-pH pH calibration curve was used to determine the pH of lysosomes lacking PQLC2-SNAP signal and the Control-pH calibration curve was used to determine the pH of PQLC2-SNAP-positive lysosomes.