Experimental model and subject details
Cell lines
HT1080 fibrosarcoma cells were from Prof Chris Marshall (ICR, UK), MDA-MB-231 breast cancer cells were from Prof Clare Isacke (ICR, UK), A375P and A375M2 cells were from Prof Richard Hynes (HHMI, MIT, USA) and WM983A and WM983B were purchased from Coriell Institute (USA). All cell lines were grown at 37oC and 10% CO2 in DMEM Supplemented with 10% FBS and 1% penicillin/streptomycin (all from GIBCO). A375P, A375M2, WM983A and WM983B were authenticated using short tandem repeat DNA profiling. All cell lines were routinely tested for mycoplasma contamination. All cell lines were kept in culture for a maximum of three to four passages and cell phenotypes were verified routinely.
For actin imaging in live cells, cells stably transfected with a LifeAct-GFP plasmid (provided by Dr Rikki Eggert, KCL, UK) were used.
Patient derived tissues
Two tissue microarrays including FFPE biopsies of 53 human primary melanomas and 45 metastasis were included in the case series (for clinical information see Supplementary Tables 1 and 2). Each biopsy was represented by two cores (1 mm diameter) from the tumour body (TB) and two cores from the invasive front (IF) areas. Tumours were classified following the most recent World Health Organization criteria. Tumour samples were processed by IRBLleida (PT17/0015/0027) and HUB-ICO-IDIBELL (PT17/0015/0024) Biobanks integrated in the Spanish National Biobank Network and Xarxa de Bancs de Tumors de Catalunya following standard operating procedures with the appropriate approval of the Ethics and Scientific Committee. Samples were collected with specific informed consent, in accordance with the Helsinki Declaration.
Animal studies
All animals were maintained under specific pathogen-free conditions and handled in accordance with the Institutional Committees on Animal Welfare of the UK Home Office (The Home Office Animals Scientific Procedures Act, 1986). All animal experiments were approved by the Ethical Review Process Committees at Barts Cancer Institute and King’s College London, in accordance with the Animals (Scientific Procedures) Act 1986 and according to the guidelines of the Committee of the National Cancer Research Institute.
Animal derived tissues
Tumours from WM983A-, WM983B-, A375P- and A375M2-EGFP cells injected subcutaneously into severe combined immunodeficient mice (SCID; CB17/Icr-Prkdcscid/IcrIcoCrl) were used for immunohistological purposes. Tumours were generated in a previous study 14.
Lung colonisation assay
For experimental metastasis assays, A375M2 cells pre-treated in vitro for 24 hours with either DMSO or Compound C (2 µM) were labelled with 10 µM CMFDA-Green (C7025, Life Technologies) for 10 min and then trypsinized and counted. 1x106 labelled cells / 0.2 ml PBS along with drugs (same concentration as pre-treatment) were injected into tail vein of NOD/SCID/ IL2Rγ-/- mice (NSG, Charles River). Mice were sacrificed 30 minutes (to confirm that equal numbers arrived at the lung) and 24 hours after tail vein injection. Lungs were extracted, washed with PBS (with calcium/magnesium) twice and fixed with 4% formaldehyde for 16 h at 4°C. Lungs were examined under a Zeiss LSM 710 Meta confocal microscope (Carl Zeiss) with a 20X objective. Data are presented as percentage of field of area covered by fluorescence, and 15 fields per mouse were analysed. n = 7 mice/condition for each experiment (2 mice sacrificed 30 minutes and 5 mice 24 hours after tail vein injection), n = 2 independent experiments.
Chemicals
Chemicals used in this study: Myosin II inhibitor blebbistatin (Calbiochem; resuspended in 95% DMSO; used at 25 µM), antimycin A (Fisher Scientific; resuspended in DMSO; used at 1 µM), oligomycin (Sigma-Aldrich; resuspended in DMSO; used at 1 µM), FCCP (Tocris Bioscience; resuspended in DMSO; used at 0.75 µM), Compound C (Sigma-Aldrich; resuspended in DMSO; used at 2 µM) and A769662 (Tocris Bioscience; resuspended in DMSO; used at 10 µM).
Antibodies
Antibodies and concentrations used: pSer19-MLC2 (#3671; 1:200 immunofluorescence; 1:50 immunohistochemistry), DDR1 (#5583; 1:1000 immunoblot, 1:200 immunohistochemistry), DRP1 (#8570; 1:1000 immunoblot), MFF (#86668; 1:1000 immunoblot), MFN1 (#14739; 1:1000 immunoblot), pThr172-AMPK (#2535; 1:1000 immunoblot; 1:150 immunohistochemistry), AMPK (#2532; 1:1000 immunoblot), MYPT (#8574; 1:1000 immunoblot), from Cell Signaling Technology; pSer172/Ser146-MFF (AF2365, 1:1000 immunoblot), pSer472-MYPT1 (AF3779; 1:1000 immunoblot), from Affinity Biosciences; GAPDH (MAB374; 1:10000 immunoblot) from Merck-Millipore; Tubulin (T6199; 1:10000 immunoblot) from Sigma-Aldrich; Integrin β1 (ab24693; 1:100 immunofluorescence), MFN2 (ab56889; 1:1000 immunoblot, 1:500 immunohistochemistry), CD44 (ab157107; 1:700 immunohistochemistry) from Abcam; OPA1 (612606; 1:1000 immunoblot, 1:500 immunohistochemistry) from BD Transduction Laboratories; Tom20 (sc-17764; 1:800 immunofluorescence) from Santa Cruz Biotechnology.
Cell culture on 3D collagen I matrices
Fibrillar bovine dermal collagen (PureCol, Advanced BioMatrix) was prepared at 1.7 mg/ml in DMEM (300 µl/well for a 24-well plate; 100 µl/well for a 96-well plate). After collagen polymerization (4 hours at 37oC – 10% CO2), cells were seeded on top in DMEM 10% FBS, allowed to adhere for 24 hours and treatments added (where appropriate). To embed cells within the collagen matrix, cells were suspended in the collagen and the appropriate volume of the suspension was seeded (100 µl/well for a 96-well plate; 250 µl/well for optical bottom 8-well µ-slide). The collagen matrices containing cells were allowed to polymerize for 4 hours at 37oC – 10% CO2. Then DMEM 10% FBS was added on top. All assays were performed with cells seeded on top of a thick layer of collagen unless otherwise mentioned.
siRNA transfection
Reverse transfection was used to transiently down-regulate the indicated genes. Two hundred and fifty thousand cells were seeded in complete media (DMEM 10%FBS) in 6-well plates with a mix containing 20 nM siGENOME SMARTpool or individual On-Target (OT) siRNA oligonucleotides (Dharmacon), Optimem-I and Lipofectamine 2000 (Invitrogen). Non-targeting siRNA were used as control. 24–48 hours after transfection cells were split and seeded for the downstream experiments/treatments. All siRNA sequences were from Dharmacon (Lafayette, USA) and are listed in Supplementary Table 3.
Immunoblotting
Cells were lysed in Laemmli Lysis Buffer and snap frozen. Then, lysates were boiled for 5 minutes, sonicated for 15 seconds and spun down. Cell lysates were resolved by SDS-polyacrylamide gels (SDS-PAGE) in non-reducing conditions and transferred to PVDF filters (0.45 µm, Immobilion™). Membranes were blocked in 5% BSA in 0.1% Tween 20-TBS. Primary antibodies were incubated overnight at 4oC. ECL Plus or Prime ECL detection System (GE Healthcare) with HRP-conjugated secondary antibodies (GE Healthcare) were used for detection. Bands were quantified using Fiji software (http://fiji.sc).
Collagen imaging
Imaging was performed with a Zeiss LSM 510 Meta confocal microscope (Carl Zeiss) with C-Apochromat X 40/1.2 NA (water) objective lenses and Zen software (Carl Zeiss). Reflectance imaging of the matrix was performed by collecting the backscattered light. Since reflectance is a surface property, extreme caution was taken to ensure the acquisition of images at the same height across all conditions.
Points of attachment
Collagen I was imaged via reflectance microscopy and points of attachment were assessed by counting the points of collagen attachment to the actin cytoskeleton (visual assessment) through consecutive z-slices. The points of attachment were manually counted using the “Cell counter” tool.
Measurement of traction stress
Live, 3D time-lapse videos coupled with image-processing were used to measure stress applied by cells within 3D collagen environments. Cells labelled with LifeAct-GFP were embedded within 3D Collagen I matrices polymerized in tissue culture wells with optical plastic bottom and imaged directly “in-well” using an inverted confocal microscope, for 8 hours. Cells were seeded such that the cellular density was sparse enough such that only one cell was present within an area of at least 250 x 250 x 50 mm in the middle of the 3D collagen matrix. This was done to ensure that the displacements measured within a certain field were only due to stress generated by the cell within the field of view.
Collagen I was imaged by collecting the reflected/backscatter light from the matrix using laser-scanning confocal microscopy 19,20,52. This approach was selected due to ease of access and lack of alterations/artificial manipulations to the matrix. By comparison of collagen I immunostained with an anti-collagen I antibody, it has been demonstrated that there is negligible detection error from reflectance imaging of Collagen I 19. The detection error arises from backscatter-negative fibrils in vertical orientation and is below 3%.
Computation of displacements and strains
Videos were generated by acquiring 3D image stacks of 30 mm thickness with step size of 1 mm. The optimal interval for acquisition of images was determined to be intervals of 2 minutes to avoid phototoxicity. The resultant videos allowed for resolution of individual collagen I fibres surrounding each cell within the x-y plane, which represents the migration of LifeAct-GFP expressing cell (mid-plane) within the surrounding matrix over 10 minutes. This set-up allowed for the optimal resolution of collagen fibres within the z-plane. The videos were then processed using a custom-written Particle Image Velocimetry (PIV) based tracking algorithm in Wolfram Mathematica 10. In order to track the matrix, 3D videos were deconstructed and analysed on a frame-by-frame basis. The deconstruction of the videos resulted in a 512x512x90 image stack corresponding to each time point. The 90 positions correspond to the interpolation along the z-axis. Superimposition of a mesh grid on the image allowed for the tracking of the position of each pixel in one frame, with respect to its relative position in the adjacent frame, at the next time point in 2D x-y plane and, in the 3D case, through each slice of the z-stack. The difference between these positions was used to calculate displacement for all three dimensions. In cases where the corresponding point in an adjacent frame could not be found, the region would be labelled zero and would then be linearly interpolated to the nearest 9 neighbouring pixels. After analysis in all three planes (xy, xz, yz) the 3D axes was then rotated and adjusted in order to reconstruct the 3D displacement data. Cell displacements calculated in X, Y and Z corresponding to each pixel in the 512x512x90 frame stacks, were converted to X, Y and Z strain values using the inbuilt derivative filter function ‘DerivativeFilter’, in Mathematica. The conversion of displacements to strains was based on the displacement-gradient technique 53. Strain data was then exported as a MATLAB file for traction stress computation in the form of a zip folder, consisting of 90 files each containing 512 variables, which in turn consisted of 512x512x3 matrices of 3D strain values.
For ease of analysis during the initial phase of development of the method, collagen displacements were measured in 2D images representing 3D projections of cells embedded within matrix with depth information incorporated. This approach allowed for the tracking of collagen displacements in 3D via maximum intensity based projections of cells in 2D.
Cell traction stress computation
In order to calculate stress from strains, the stiffness of the bovine collagen matrix was calculated using AFM microscopy. Stiffness of bovine collagen was ~ 19 Pa, comparable to the stiffness observed in other studies 19,54. Stress were then calculated by integrating the strain data, the directional information, depth information and stiffness of the matrix. A custom algorithm was written in MATLAB, using a ‘direct Traction Force Microscopy (TFM)’ method 55, and stress were directly calculated from computed strains, as opposed to more complicated and computationally extensive methods involving the solution of inverse equations and boundary conditions. The mathematical approach was based on that used by 18, following the assumption of a linearly isotropic material or substrate. According to this assumption, the Cauchy relation for traction stress, τ, is:
τ = δ.n Eq. (1)
where ‘δ’ is the Cauchy stress tensor and ‘n’ the direction of the normal vector.
The MATLAB code was designed to process each of the aforementioned 512 variables, setting the X, Y and Z strain values as the principal strains (diagonal elements) of a 3D symmetric strain tensor ‘ε’, defined as:
ε = \(\left[\begin{array}{ccc}{\epsilon }_{11}& {\epsilon }_{12}& {\epsilon }_{13}\\ {\epsilon }_{21}& {\epsilon }_{22}& {\epsilon }_{23}\\ {\epsilon }_{31}& {\epsilon }_{32}& {\epsilon }_{33}\end{array}\right]\)Eq. (2)
Equation (3) below was solved in order to obtain the shear modulus ‘µ’, which was then input into Eq. (4), to obtain the stress tensor ‘δ’, for which isotropic linearly elastic properties are assumed 54. The constitutive mechanical properties required for these calculations included a Young’s modulus ‘Ε’ of 28 Pa, measured for our collagen matrix, and a Poisson ratio ‘v’ of 0.25 based on that measured by Steinwachs et al (2016).
Ε = 2µ (1 + v) Eq. (3)
δ = 2µε Eq. (4)
The MATLAB function ‘eig’ was then called to solve the eigen Eq. (5) for the three eigenvalues (λ1, λ2, λ3) and eigenvectors (V1, V2, V3) of this stress tensor. The eigenvalues correspond to the traction stress solutions of the stress tensor ‘δ’, whilst the eigenvectors correspond to the direction of the normal ‘n’, relative to the surface on which the traction stress is acting.
Ε = (δ-λI) V Eq. (5)
where ‘Ε’ represents the set of vectors which satisfy (δ - λl) V = 0.
The matrix dot product of the stress eigen values with their correct and corresponding eigen vectors, yields the traction stress ‘τ’, in the Cauchy relation (Eq. 1). This step is more complicated in 3D, compared to 2D, as there are 9 different eigen vector permutations and therefore combinations of the eigen vectors with their respective eigen values.
Appropriate considerations therefore had to be incorporated into the code in order to deal with this difficult step whilst being able to process the data efficiently.
Once the traction stress had been computed in X, Y and Z (Τx, Τy, Τz), the magnitude of these stress was computed using the standard vector combination relation:
\(\left|\text{T}\right| = \sqrt{{T}_{x}^{2}+{T}_{y}^{2}+{T}_{z}^{2}}\) Eq. (6)
By this stage, one data set would consist of 90 individual 512x512 matrices of traction stress magnitudes corresponding to each pixel. Before generating a visual representation, it was important to vertically flip and rotate this matrix anticlockwise by 900 in order to correct for the default image conversion adjustments made by MATLAB and Mathematica software, respectively. This also ensured accurate analysis of cell movements later on, when cell outlines were overlaid on the maps. After processing each data stack, a 512x512 pixel colour map was generated for each slice in the stack using a ‘jet’ LUT, intrinsic to MATLAB. According to this colour scheme, the resulting gradient based stress maps were colour coded such that the red areas of the reaction maps correspond to traction stress while blue regions correspond to minimal or zero traction stress and are found in areas of matrix where no substantial impact due to cell movement was detected.
Cell morphology on collagen
Cell morphology of cells seeded on collagen matrices was quantified on still phase-contrast images using ImageJ. Cell morphology was assessed using the morphology descriptor tool “roundness” after manually drawing around the cell. Values closer to 1 represent rounded morphology; values closer to 0 represent more spindle-shaped cells.
Cell adhesion to collagen I
Collagen matrices were prepared in 96-well plates. Ten thousand cells/well were seeded in duplicate or triplicate in the 96-well plates and incubated at 37oC for the indicated times. The centre of the well was then imaged with a phase contrast microscope, washed three times with PBS and imaged again with the same settings. Each well was imaged before and after washing. Cells were counted manually using “Cell counter” function in ImageJ and results are presented as percentage of adhered cells, calculated as number of cells after washing divided by the number of cells before washing.
3D invasion
For 3D invasion assays, cells were resuspended in serum-free bovine collagen I solution at 2.3 mg/ml to a final concentration of 14 000 cells per 100 µl of matrix and spun down, in a 96-well plate. After the matrix was polymerized, 10% FBS-containing media was added on top of the matrix. After 24 hours cells were fixed, stained with Hoechst and imaged using a Zeiss LSM 710 confocal microscope. Invasion was calculated as number of invading cells at 50 µm divided by the number of cells at the bottom.
Immunofluorescence and confocal imaging
Cells seeded on top of collagen matrices were fixed with 4% formaldehyde for 15 minutes at room temperature. Then, cells were permeabilised for 20 minutes using 0.3% Triton X-100 in 5% BSA-PBS, blocked in 5% BSA-PBS for 30 minutes and immunostained with primary antibody overnight at 4oC. Next, samples were incubated with Alexa Fluor 546-phalloidin and Alexa Fluor 488 or 647 secondary antibodies for 2 hours at room temperature. Nuclei were stained with Hoechst prepared in PBS. Antibodies were diluted in 5% BSA-PBS.
Images were taken with a Zeiss LSM 510 Meta confocal microscope with Plan-Apochromat 40x/1.2 NA (water) objective lenses, Zeiss LSM 710 confocal microscope with Plan-Apochromat 40x/1.3 Oil DIC M27 and Zeiss LSM 880 confocal microscope with Airyscan superresolution mode and Plan-Apochromat 63x/1.4 Oil DIC M27 objective lenses (Carl Zeiss, Germany). Zen software was used to acquire images (Carl Zeiss, Germany). Images were analysed using ImageJ software (NIH). For pMLC2 quantification, fluorescence signal was quantified by calculating the area occupied by pMLC staining in single cells relative to the cell area. For pMLC2 distribution, line scan analysis was performed in ImageJ.
Mitochondrial mass and mitochondrial activity
Mitochondrial activity and mitochondrial mass comparing adherent versus floating cells were analysed by flow cytometry. Cells were seeded either on regular cell culture plates or polyhema pre-coated plates (for non-adherent conditions). After 24 hours cells were collected, spun down and resuspended with a solution containing MitoTracker Deep Red (25nM) and TMRE (200nM), at 37oC for 20 minutes. After, FACS buffer (1% BSA, 2mM EDTA and 0.1% NaN3 in PBS) was added to samples and immediately analysed on a BD FACS CANTO II flow cytometer. Data was analysed using FlowJo software (Tree Star).
Mitochondrial activity and mitochondrial mass comparing cells on top of collagen were analysed by confocal imaging. Live cells expressing LifeAct-GFP seeded on top of a collagen I matrix were incubated with MitoTracker Deep Red (25 nM) for mitochondria visualisation and with tetramethylrhodamine ethyl ester (TMRE; 200 nM) for mitochondrial activity analysis, at 37oC for 20 minutes. Then, were kept in FluoBrite DMEM for imaging. Z-stack images were taken with a Nikon Eclipse Ti Inverted equipped with a Yokogawa CSU-X1 Spinning Disk unit 100x oil objective lenses. Images were analysed using ImageJ software (NIH). Mitochondrial activity (TMRE signal) was quantified from maximal intensity projections and normalized by mitochondrial mass (MitoTracker Deep Red signal).
Mitochondrial morphology
For mitochondrial circularity analysis, MitoTracker Deep Red z-stacks images were imported in to Mathematica 11, converted to 3D and decomposed in to frequency bands using difference of Gaussian filters using radii ranging from 1-to-32 in powers of 2. This enabled simultaneous smoothing and background subtraction. The frequency-decomposed images were then combined to create a frequency-encoded image. The images were then Top-hat transformed with a radius of 4 to highlight the mitochondria and reduce uneven intensities. Filtered images where then thresholded and binary objects that were less than ten pixels in size were deleted. The internal Mathematica function “Component Measurements” was then used to calculate the circularity of each object of the image. These were then exported as “csv” files for further analysis.
For the analysis of mitochondrial branches per cells, a semi-automated image-based analysis was performed in ImageJ using Mitochondria Analyzer 56. Single plane images of Tom 20 fluorescently-labelled mitochondria in cells grown on a collagen I matrix were pre-processed and converted to 8-bit images. Next, 2D Optimize Threshold command was used to identify the appropriate settings on test samples from each set acquired under similar imaging conditions. Block size of 2.25 µm and C-value of 28 was used for the 2D Analysis in batch mode. Analysis was obtained on a per-cell basis.
NMR metabolomics
The metabolic profiles were assessed using NMR metabolomics. Three million cells were seeded in 14.5 cm dishes 48 hours prior to dual-phase metabolite extraction. Media was removed and cells were washed twice with cold PBS. Then, each dish was placed on dry ice for 2 minutes to quench the metabolome. Afterwards, 2.2 mL of cold methanol was added and cells were scraped and transferred to a pre-cooled 15 mL tube. 2.2 mL of cold chloroform was added to each tube and mixed in a tube rotor for 10 minutes at 4oC. Then, 2.2 mL of milli-Q water was added to each tube and the contents were mixed by inversion and incubated on ice for 10 minutes to allow the formation of a stable bilayer. Samples were then centrifuged at 4oC and 1550 rpm for 45 minutes to separate the top layer and a bottom layer, containing metabolites soluble in the polar and apolar fraction, respectively. The polar fraction was dried in an Eppendorf concentrator (Concentrator plus, Eppendorf).
Prior to NMR experiments, samples were resuspended in a 90/10 H2O/D2O buffer, with 100 mM Na2HPO4, 5 mM TSP and 4 mM NaN3. NMR spectroscopy was performed on a Bruker Avance NEO 600 MHz NMR spectrometer equipped with a TCI Prodigy CryoProbe (Bruker). Spectra were acquired at 298 K and consisted for each sample of a 1D 1H PURGE spectrum 57, which was then phase corrected, baseline corrected and the chemical shifts were referenced to the TSP peak at 0.0 ppm.
Spectra were aligned by the PAFFT method (peak alignment by fast Fourier transform) and then normalized by probabilistic quotient normalization (PQN) 58. To determine whether spectra cluster into groups, PLS-DA with k-fold cross-validation were performed. PLS-DA of the full 1D 1H spectrum, after alignment, normalization using PQN, and log-scaling of the spectra was done. For multivariate analyses, spectra were scaled by the pareto method, which decreases the influence of high intensity peaks while emphasizing low intensity signals, such that both high and low intensity signals have equal significance in the model 59. 1H-NMR profiling of these cellular extracts allowed the identification of the main compounds present in the mixture. The metabolite assignment was done by comparing peak chemical shifts to those found in literature, in Human Metabolome Database (HMDB) and in the Biological Magnetic Resonance Bank (BMRB). Furthermore, all the metabolite structures were confirmed by 2D NMR experiments. Relative quantification of metabolites was achieved by integration of peaks to calculate peak area, which is proportional to the number of nuclei that give rise to the signal. Mean ± SD of peak areas were calculated for each metabolite.
Metabolic assays
ECAR and OCR measurement
In 2D systems, oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) were measured performing a Mitochondrial stress test and a Glycolysis stress test respectively, using a Seahorse XFe96 analyser (Agilent), following manufacturer instructions. Briefly, 5 000 cells were seeded on Seahorse XF96 cell culture microplates in DMEM 10% FBS and incubated overnight at 37oC and 10% CO2. Next, media was changed to XF DMEM media, supplemented as indicated by the manufacturer and cells were incubated at 37oC without CO2 for 1 hour, before proceeding immediately to perform the OCR or ECAR analysis. OCR was monitored following the sequential addition of oligomycin (1 µM), FCCP (0.25 µM) and rotenone/antimycin A (0.5 µM). ECAR was monitored following the sequential addition of glucose (10 mM), oligomycin (1 µM) and 2-deoxy-D-glucose (2DG, 50 mM).
For the measurement of OCR in 3D cell cultures, MitoXpress Xtra Oxygen Consumption assay (Agilent) was used. Cells were seeded embedded in a 3D collagen matrix as explained above. Twenty-four hours later, media was replaced with 60 µl DMEM 1% FBS containing MitoXpress Xtra reagent. Then, wells were sealed with HS mineral oil and time-resolved fluorescence (TR-F) was measured immediately for 2 hours, using a BMG Omega plate reader, following manufacturer instructions. Glucose oxidase (0.1 mg/ml) was used as a cell-free positive control. Antimycin A (1 µM) and FCCP (0.75 µM) were used as cell-based negative and positive control, respectively. Slope between 30 and 90 minutes was used to quantify OCR, following manufacturer’s protocol.
Cellular ATP measurement
Luminescent ATP detection assay
For the measurement of total ATP in A375P cells, the luminescent ATP detection assay (ab113849, Abcam, Cambridge, UK) was used. Briefly, the day before the assay 5 000 A375P cells/well were seeded on untreated and polyhema-treated (P3932, Sigma-Aldrich) 96 well plates and allow to grow overnight in DMEM 10% FBS, incubated at 37oC and 10% CO2. A375P cells cultured on polyhema plates were grown as suspension cells. The day after, ATP was quantified according to the manufacturer’s protocol.
Seahorse XF Real-Time ATP Rate Assay
For the quantification of ATP production rate from both glycolytic and mitochondrial pathways, Agilent Seahorse XF Real-Time ATP Rate Assay was performed according to the manufacturer’s instructions (Agilent Technologies, Santa Clara, CA) for adherent and floating cells. Briefly, the day prior the assay, 5 000 A375P cells/well, previously grown under adherent conditions, were seeded on a Seahorse XF96 cell culture microplate in DMEM 10% FBS, incubated at 37oC and 10% CO2. On the day of the experiment, media was changed for XF DMEM (pH = 7.4) supplemented with 1 mM pyruvate, 2 mM glutamine and 10 mM glucose (Agilent, USA). Furthermore, 7 500 A375P cells, previously grown under floating conditions for 24 hours, were seeded on the Seahorse XF96 cell culture microplate in the XF media specified above. Both adherent and floating cells were then incubated at 37oC without CO2 for 1 hour. During this incubation period, oligomycin and rotenone/antimycin A (Agilent, USA) were prepared in Seahorse media to achieve final concentrations of 1.5 µM and 0.5 µM respectively when injected. The glycolytic and mitochondrial ATP production rate were calculated according to manufacturer instructions. The total ATP production rate is the sum of the glycolytic and mitochondrial ATP production rates.
Finally, the ratio between mitochondrial ATP and glycolytic ATP production rate as well as the ratio between mitochondrial ATP and total ATP production were represented.
Measurement of ATP:ADP ratio in 3D
PercevalHR lentivirus were generated in HEK293T cells. Media with lentiviruses was added to recipient cell lines HT1080, A375M2 and A375P. PercevalHR expressing A375P cells were then transfected with DDR1 siRNA following the protocol as explained above. After 48 hours cells were trypsinized, spun down and re-suspended in collagen I. Two hundred and fifty microliters of collagen-cell suspension were added per well in optical bottom 8-well µ-slide (Ibidi). Collagen was allowed to polymerize for 4 hours at 37oC and DMEM 10% FBS was added on top. Twenty-four hours later, media was replaced by FluoBrite DMEM for imaging. Perceval HR expressing cells were sequentially excited using 405 and 488 nm lasers with emission collected at 540 nm for both channels 21,60. Z-stack images for the entire volume of the cell were taken using Zeiss LSM 880 with Fast Airyscan mode, Plan-Apochromat 63x/1.4 Oil DIC M27 objective lenses and Zen software (Carl Zeiss, Germany). Pixel-by-pixel ratio of ATP bound (488) / ADP bound (405) signals were calculated using ImageJ RatioPlus tool.
Cell viability
Propidium iodide was used to stain dead cells. After spinning down spent media and cells, cells were resuspended in a solution of Propidium iodide (10 µg/ml) in PBS and incubated for 15 minutes on ice. Immediately afterwards, samples were analysed on a BD LSR Fortessa flow cytometer. Data was analysed using FlowJo software.
Immunohistochemistry
Paraffin-fixed sections were sequentially stained for DDR1, MFN2, OPA1, pMLC2, pAMPK and CD44, as previously described 15,61. Tumour tissues were formalin-fixed and paraffin embedded as per standard protocols. Sections of 4 µm thickness were heated at 60oC for 1 hour and then incubated in xylene and ethanol series, with 2x 5 minutes H2O2/ethanol incubations to block endogenous peroxidase. Antigen retrieval was performed in Antigen Unmasking Solution pH 6 (H-3300, Vector Labs) using a pressure cooker system (110oC for 10 minutes). Samples were washed in Dako Wash Buffer (S3006), before primary antibody incubation (40 minutes, 1:200 anti-DDR1, CST #5583), diluted in Antibody Diluent Reagent Solution (003218, invitrogen/ThermoFisher Scientific). Samples were washed and incubated with ImmPRESS® polymer secondary goat anti-rabbit antibody (goat anti-rabbit, RTU, Vector Labs, MP-7451) for 45 minutes. The reaction was developed using VIP peroxidase substrate solution (Vector Labs, SK-4600) for 10 minutes. All incubations were carried out at room temperature. Slides were counterstained with haematoxylin and mounted using DPX mounting medium (06522-500ML, sigma). Slides were imaged using the NanoZoomer S210 slide scanner (Hamamatsu, Japan). The next day, slides were processed using the same procedure. Previous staining was stripped through the antigen retrieval step. In the second round, an anti-MFN2 primary antibody was used (40 minutes, 1:500, ab56889) and ImmPRESS® polymer secondary goat-anti mouse antibody (40 minutes, RTU, Vector Labs, MP-7452); developing, mounting and imaging was performed as before. The third round was performed as above, where slides were incubated with an anti-OPA1 antibody (40 minutes, 1:500, BD-612606) and ImmPRESS® polymer secondary goat-anti mouse antibody (40 minutes, RTU, Vector Labs, MP-7452) was used; developing, mounting and imaging was performed as before. In the forth round, anti- pMLC2 (ser19) antibody was added to the panel (40 minutes, 1:50, CST #3671) and ImmPRESS® polymer secondary goat-anti rabbit antibody (40 minutes, RTU, Vector Labs, MP-7451) was used; developing, mounting and imaging was performed as before. In the following fifth round, pAMPK antibody (40 minutes, 1:150, CST#2535) and ImmPRESS® polymer secondary goat-anti rabbit antibody (40 minutes, RTU, Vector Labs, MP-7451) were used; developing, mounting and imaging was performed as before. Finally, the sixth round included CD44 antibody to stain cell membranes which allowed the quantification of cell morphology. Tissue sections were incubated with anti-CD44 (40 minutes, 1:700, ab157107) and ImmPRESS® polymer secondary goat-anti rabbit antibody (40 minutes, RTU, Vector Labs, MP-7451) was used; developing, mounting and imaging was performed as before.
Image Analysis
Staining quantification was performed using QuPath 0.1.2 (Bankhead et al., 2017). For each marker, whole section images (WSI) from mouse tissue and TMAs were analysed performing positive cell detection, and three different thresholds were applied according to the intensity scores (0, 1, 2 and 3). Next, the software was trained by creating a random tree classification algorithm combined with the intensity information, in order to differentiate tumour from stroma, necrosis and immune cells. Then, IHC staining was graded semiquantitatively by considering the percentage and intensity of the staining. A histologic score (Hs) was obtained from each sample, and values ranged from 0 (no immunoreaction) to 300 (maximum immunoreactivity) arbitrary units. The score was obtained by applying the following formula, Hs = 1 × (% light staining) + 2 × (% moderate staining) + 3 × (% strong staining). For co-localization analysis, images for the 5 markers markers (DDR1, MFN2, OPA1, pMLC2 and pAMPK) were aligned in FIJI v1.52p using TrackEM2 module. Next, colour deconvolution was performed using AEC-Haematoxylin vectors and a composite was created using channel-2 (red) for each staining. The composite was adjusted inverting the LUT for each marker and given a pseudocolour. We used QuPath DoG superpixel segmentation to quantify cell morphology. CD44 staining was used to perform an accurate watershed segmentation. Finally, shape measurements were added in QuPath such as rounding in all the detections. To calculate the amoeboid score (As) the same ROI used in CD44 for each case we used with pMLC2 staining and the intensity for such marker was calculated in each detection. The As was calculated combining roundness and pMLC2 intensity for each detection, as shown in the formula: As = roundness (0–1) * Hs pMLC2 (0-300) = 0-300
Values for As range from 0 to 300 were 0 represents a very elongated cell (roundness = 0) with null levels of Myosin II (Hs = 0), while the representative amoeboid cell harbours As of 300. For each case values are represented as mean of all detection per ROI.
Quantification and statistical analysis
Statistical analyses were performed using GraphPad Prism 8 software (GraphPad, San Diego). The following statistical analysis were used: two-sided Student’s t-test, Mann-Whitney, Wilcoxon test, two-sided one-way ANOVA with Tukey’s or Dunnett’s post hoc test and two-way ANOVA with Dunnett’s post hoc test. Data were represented in bar graphs and dot plots as mean ± standard error of the mean (SEM) and box plots as median (centre line), interquartile range (box) and min-max values (whiskers). In general, experiments were carried out at least 3 independent times with 2–3 technical replicates, unless otherwise stated. Significance was defined as p < 0.05. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.
MATERIALS AVAILABILITY
The code generated during this study will be available at GitHub.
Supplementary Data 1. Script for 3D traction stress calculation.
All unique/stable reagents generated in this study are available with a completed Materials Transfer Agreement.