Animal studies
For removal of Pik3ca- floxed allele, Pik3caRBD/lox and Pik3caWT/flox mice64 were given 3.2 mg tamoxifen (Sigma) dissolved in 80 μl of corn oil by oral gavage once per day during 3 consecutive days. Efficiency of tamoxifen treatment was routinely performed by genotyping for the presence of the floxed allele.
For paw oedema studies Pik3caRBD/- and Pik3caWT/- mice64 were divided into groups of four two weeks before edema induction. Before inducing the paw edema, the mice were anesthetized with 4% isofluorane. To induce the edema, mice received ipsilateral i.pl. injection (30 µl) of either zymosan (10 μg/μl, Sigma Aldrich) or PBS into the back-hind paw. Injection of 0.1mg/kg Buprenorphine (NOAH, Vetergesic) was given for pain prevention. Paw thickness was measured using a caliper every hour during the first 6 hours after injection and then at 8, 10 hours and twice per day afterwards. Buprenorphine was injected twice per day during the length of the experiment. Mice were kept, managed, and sacrificed in the NUCLEUS animal facility of the University of Salamanca according to current European (2007/526/CE) and Spanish (RD 1201/2005 and RD53/2013) legislation. All experiments were approved by the Bioethics Committee of the Cancer Research Center.
Isolation, culture and treatments of BMDM
Bone marrow cells from tibias and femurs of 12-14-week-old Pik3caRBD/Lox mice and Pik3caWT/Lox littermates were cultured with DMEM supplemented with 10% FBS, 100 units/ml penicillin, 100 μg/mL streptomycin, 2 mM L-Glutamine and 20 ng/mL M-CSF for 7 days. 4-hydroxytamoxifen (Sigma Aldrich) (100 nM) was added to culture media on day 3 to eliminate Pik3ca-Lox allele. The differentiated BMDM were then detached using cell dissociation buffer (C5914-100, Merck) and cultured in DMEM supplemented with 10% FBS, 100 μg/mL streptomycin, 2 mM L-Glutamine, 20 ng/mL M-CSF for unstimulated BMDMs. For macrophage polarization towards an inflammatory phenotype 20 ng/ml IFNγ (Peprotech) and 100 ng/ml LPS (Sigma Aldrich) was added to culture media.
Generation of chimeric animals
Chimeric mice exhibiting WT or RBD deficient leukocytes were generated by lethal irradiation with 5.5 Gy twice, 4 h apart of LysM GFP recipient animals (mice exhibiting endogenously GFP fluorescent monocytes and neutrophils) followed by an injection of bone marrow cells (1.5 × 106 cells/recipient i.v.) from C57BL/6 WT or RBD donor mice. Chimerism was then assessed 4 weeks later by flow cytometry from blood samples (reconstitution of 99.8 ± 0.2 % and 99.8 ± 0.1% for WT and RBD deficient donor cells, respectively; n =5 mice per group).
Neutrophil depletion
Neutrophil depletion of chimeric mice was induced by intraperitoneal injection of anti-GR1 25μg/mouse/day for 3 days). Numbers of blood circulating monocytes and neutrophils were quantified by flow cytometry pre- and post-depletion. Neutrophils were found to be reduced by 99.5%, whilst this anti GR1-depleting protocol had no effect on blood monocyte proportion (n=5 mice/group).
Brightfield intravital confocal microscopy
Mesenteric inflammation was induced following intraperitoneal injection of mouse recombinant CCL2 (500ng/mouse in 500uL of PBS). Six hours later, anesthetized chimeric mice (150 mg/kg ketamine, 7.5 mg/kg xylazine, i.p.) were placed in supine position on a heating pad (37 ºC) for maintenance of body temperature. The mesenteric vascular bed was exteriorized, placed on a purpose-built stage of an upright brightfield microscope (Zeiss Axioskop). Mesenteries were superfused with warmed (37 ºC) Tyrode’s solution (Sigma). After a 5-min equilibration period, analysis of leukocyte-endothelium interactions was made in at least 9 (and up to 16) randomly selected segments (100 µm in length) of post-capillary venules (20–40 µm in diameter) for each mouse. Leukocyte rolling was quantified by counting the number of rolling cells passing a fixed transversal line in the middle of the vessel segment for 5 min. Leukocyte adhesion (stationary position of the cell for 30 s or longer) was quantified along a 100 µm vessel length and data were normalized as the number of cells per 500um vessel segments. Leukocyte extravasation response was quantified within 50 µm on either side of the 100 µm vessel segment in the perivenular tissue; and data were normalized as the number of extravasated leukocytes per mm2 of extravascular tissue. At the end of the analysis period, mice were humanely killed by cervical dislocation.
Immunofluorescence
BMDM were fixed using 4% paraformaldehyde, permeabilized with 0.1% Triton X-100 and blocked for 1 h with 3% BSA in PBS before incubation with the primary antibodies, used at a 1:100 dilution: Lamp1 (#553792, BdPharmigen), Deoxyribonuclease I-Alexa Fluor™ 488 Conjugate (#D12371, Invitrogen, 1:2000). To stain actin cytoskeleton, Alexa Fluor™ 647 Phalloidin (Invitrogen, 1:10 000) was directly added to the primary antibody mixture. Alexa Fluor 488- or Alexa Fluor 555-conjugated secondary antibodies (Invitrogen) were used to detect the indicated proteins at a 1:1000 dilution. Cells were counterstained with DAPI on the mounting solution (ProLong Gold Antifade Reagent with DAPI, Invitrogen). Images were taken using a Zeiss LSM510 confocal microscope or Leica DM6 B THUNDER Imager 3D Tissue.
Transwell migration assay
Transwell migration assays were carried out using the 6.5 mm Transwell® with 8.0 µm Pore Polyester Membrane Insert (Corning). 9x104 MEFs from wild type mice were used as a chemo-attractant to encourage macrophage migration. 8x105 BMDM were seeded in the transwell. Transwells were performed following the manufacturer instructions.
Random migration assay
For random migration assays, BMDMs were seeded in 24-well plates coated with matrigel (0.5mg/ml) and labelled using CellTracker™ Red CMTPX Dye 1 μM (ThermoFisher) for 30 minutes. 24 hours later LPS + IFN‑γ was added when necessary. Triplicates of each condition and genotype were prepared. Time-lapse imaging was carried out for 24 h. One image was taken every 10 min within the same well using a Nikon microscope driven by Metamorph (Molecular Devices, Chicago, IL, USA). A total of 80-100 cells per condition were tracked using the Fiji plugin Trackmate. Pre-processing was done using Mexican hat filter 3.0 radius to increase particle detection. Images were segmented using the fluorescence channel with the Laplacian of the Gaussian detector with a 30 μm estimated particle diameter, a 10.0 threshold and median filter option selected. Segmented objects were linked from frame to frame with a Linear Assigment Problem (LAP) tracker with 45 μm frame-to-frame linking distance and 2 frame gap closure. Criteria for track acceptance were track duration at least the 90% of the video. Tracks were visually inspected for completeness and accuracy of the tracking.
Secretome mass spectrometry
Samples for secretome analysis were prepared as previously described130. In brief, 100ug of proteins were digested into peptides using trypsin and peptides were desalted using Oasis HLB extraction cartridges (Waters UK Ltd)) and eluted with 50% acetonitrile (ACN) in 0.1% Trifluoroacetic acid (TFA).
Dried peptides were dissolved in 0.1% TFA and analysed by nano ACQUITY liquid chromatography (Waters Corp., Milford, MA, USA) coupled on-line to a tandem LTQ Orbitrap XL, mass spectrometer (Thermo Fisher Scientific)131. Gradient elution was from 5% to 25% buffer B in 180 min at a flow rate 300nL/min with buffer A being used to balance the mobile phase (buffer A was 0.1% formic acid in water and B was 0.1% formic acid in ACN). The mass spectrometer was controlled by Xcalibur software and operated in the positive mode. The spray voltage was 1.95 kV and the capillary temperature was set to 200 ºC. The LTQ Orbitrap XL was operated in data dependent mode with one survey MS scan followed by 5 MS/MS scans. Label-free quantitative proteomics analysis was performed using three independent biological samples per group. Additionally, each sample was analysed in technical duplicates. To ensure robust quantitative analysis, we utilized LTQ Orbitrap XL tandem mass spectrometry (MS/MS) to generate six distinct mass spectral profiles from each group.
MS raw files were converted into Mascot Generic Format using Mascot Distiller (version 2.3.0) and searched against the SwissProt database (release December 2015) restricted to human entries using the Mascot search daemon (version 2.3.1). Allowed mass windows were 10 ppm and 600 mmu for parent and fragment mass to charge values, respectively. Variable modifications included in searches were oxidation of methionine, pyro-glu (N-term) and phosphorylation of serine, threonine and tyrosine.
Spectral counting quantification method relies on the number of times peptides are identified by tandem mass spectrometry (with expectancy value <0.05) from a given protein. Spectral counts were obtained from Mascot result (DAT) files using a python script written in house in the Mascot Parser Toolkit environment (version 2.4.x).
Proteomic data analysis
The proteomic data obtained consisted of 6,844 peptides and 30 samples: Pik3caWT/− and Pik3caRBD/− BMDMs in steady state conditions (labelled as cell samples: WT/- and RBD/-), phagocytosing apoptotic cells (labelled as cell samples: WT/-Phag and RBD/-Phag), and the apoptotic LKR10 cells alone (labelled as LKR). For each of these samples, proteomic experiments were performed with 6 replicates (3 biological replicates x 2 technical replicates), yielding a data set of 30 samples.
The first analytical step was to remove all peptides for which there was no information contained in the proteomic raw data matrix and the peptides for which 85% or more of the signal values were missing. All these peptides were specific of mouse proteins and in many cases were unique. The corresponding proteins were annotated and labelled together with each measured peptide. Next, low-quality samples were also removed, testing the overall signal per sample to identify if there were clear outliers with very low signal or with a very different signal distribution. Comparison of the overall signal distributions of the 30 samples (comparing boxplots) and identified 3 samples that were very different were obtained and discarded (WT/-Phag_s3r2 (sample 3, replicate 2), RBD/-_s2r1 and LKRc_s1r1). These 3 samples showed a median signal in their distributions that deviated >20% from the median signal of the distributions of all other samples.
Differential expression analysis for each peptide of each protein were next performed. The algorithm used to carry out this analysis was limmaVoom within EdgeR R package65, 66. Prior to this analysis, a Bartlett test was performed to see the homogeneity of variances, verifying that for this data we cannot consider equality of variances and this factor was included in the differential expression algorithm. With this algorithm, normalization factors to use a-posteriori were calculated and, transformation and calculation of the variance weights was performed. The model to fit before using Voom as specified since it uses the variances of the model's residuals (observed - fitted). Finally, an estimation of the contrast for each feature tested (i.e. each peptide) was carried out using the Empirical Bayes approach in limma as previously described67. Peptides were ordered by the p.value of the limma test considering significant peptides changed only with a p.value below 0.05 and with a log2(Fold-Change) >|2|. All these analyses were performed using the statistical computing language R and packages or libraries obtained from R-cran (cran.r-project.org) or Bioconductor (www.bioconductor.org).
Cytoscape software (v3.9)68 including GeneMania app69 was then used to generate and visualize protein-protein networks of the significantly altered proteins selected in secretome analysis of unstimulated and phagocyting BMDMs. This tool provides information on protein-protein associations based in co-expression studies and also based in physical interaction studies.
Western blot analysis
Immunoblot was performed per a general western-blot protocol (Abcam). Total protein was extracted using Cell Lysis Buffer (Cell Signaling Technology) supplemented with c0mplete mini protease inhibitor cocktail (Roche), 50 mM sodium fluoride and 1 mM of PMSF. Protein was quantified using Bradford Method (Bio-Rad). 20 μg of protein was separated by SDS-PAGE and transferred to 0.2 um pore-size PVDF membranes (Sigma-Aldrich). Blots were probed using the following antibodies, at a concentration 1:1000 unless otherwise stated: phopho-LIMK1 (Thr508)/LIMK2 (Thr505) (#3841, Cell signalling), cofilin (sc-376476, Santa Cruz Biotechnology), phospho-cofilin (Ser3) (#3311, Cell Signalling), cathepsin B (12216-1-AP, Proteintech), cathepsin D (21327-1-AP, Proteintech), α-tubulin (ab15246, Abcam; concentration 1:5000). Horseradish peroxidase-conjugated secondary antibodies (Amersham) were used (1:5000) and detected using an enhanced chemiluminescent substrate (Amersham). Signal was detected using an iBright 1500 System (Invitrogen).
Flow cytometry analysis
Single-cell suspensions from cultured cell, spleen or blood monocytes were generated from mice, washed twice in staining buffer and incubated with 1:100 Fc-block (BD Biosciences, #553142) diluted in FACS buffer. Cells were subjected to surface antibody staining with labelled antibodies diluted in staining buffer for 30 min at 4 °C: CD3-PE-Cy7 (#100328, Biolegend), CD4-BV605 (#100548, Biolegend), CD8-APC (#100712, Biolegend), CD19-PerCP-Cy5.5 (#115534, Biolegend), CD45-BV785 (#103149, Biolegend), Ly6C-PerCP-Cy5.5 (#128012, Biolegend), Ly6C-E450 (#48-5932-82, eBioscience), Ly6G-AF700 (#56-5931-82, eBioscience), CD11b-BV650 (#101239, Biolegend), F4/80-PE-Cy7 (#123114, Biolegend), F4/80-PE (#123110, Biolegend), CCR2 (CD192)-PE-Vio ® 770 (#130-108-724, Miltenyi Biotec). After incubation, cells were washed in staining buffer and analysed immediately. For all staining, isotype controls were used.
Samples were acquired on a BD LSR FORTESA FACS or FACS Aria III machine that uses FACS DIVA software (BD Biosciences). Compensation was performed using 1 drop of Ultracomp ebeads (eBioscience) in 300μl of FACS buffer. 1μl of each antibody used in the pool was mixed with 100μl of compensation beads solution and acquired. Analysis was performed with FlowJo software (FlowJo V10.4). Once the different pools were compensated samples were acquired.
Phagocytosis assay
For phagocytosis assay, BMDMs in suspension in DMEM without FBS were labelled with 1:200 red cell tracker (Molecular Probes) for 30 minutes at 37°C. Cells were then centrifuged for 5 minutes at 300g at 4°C, supernatant was removed and labelled BMDMs were washed twice with 5ml of PBS and plated overnight in complete DMEM containing 20 ng/ml M-CSF. On the following day, 100 ng/ml LPS (Sigma Aldrich) was added to BMDMs overnight.
LKR10 cells (murine lung cancer cell line) were stained with red cell tracker (Molecular Probes) as previously described for macrophages. Stained cells were plated in complete DMEM medium and, after 12 hours, 50 μM Cisplatin (MCE MedChemExpress) was added to the media and left overnight. BMDMs were then incubated with apoptotic cancer cells at a 1:2 ratio and cultured at 37 °C for different time points in DMEM supplemented with 10% FBS.
Flow cytometry data were acquired using a BD LSR FORTESSA FACS instrument with FACS DIVA software (BD Biosciences) and analysed using FlowJo V10.4 software. A minimum of 2x105 events were acquired and analysed. Data analysis and interpretation was done using FlowJo software (FlowJo V10.4).
Image analysis
Confocal images were post-processed and analysed using Fiji distribution of ImageJ version 1.53q. Cell shape descriptors such as “aspect ratio” (AR), “circularity” (C) and “cell area” were measured using Fiji. Specifically, aspect ratio is calculated as (major axis×minor axis−1) therefore representing solely the degree of elongation, whereas circularity is calculated as [4π*(area × perimeter−2)], thus representing the degree of similarity to a circumference with a value ranging from 0 to 1 (perfect circle).
Histology
Tissue was fixed using 4% formaldehyde for 48h, dehydrated and paraffin-embedded. Sections (3 μm) were cut and stained using hematoxylin-eosin. For immunodetection, citrate pH 6 buffer was used for antigen retrieval. Staining was used using the following primary antibodies: CD68 (ab125212, Abcam, 1:200), cathepsin B (12216-1-AP, Proteintech, 1:100), cathepsin D (21327-1-AP, Proteintech, 1:400). Dako EnVision+ System HRP labelled Polymer secondary antibodies (Dako) were used, and DAB+ Substrate Chromogen System (Dako) was used for color development.