Waiting
Login processing...

Trial ends in Request Full Access Tell Your Colleague About Jove

Cancer Research

A 3D Spheroid Model as a More Physiological System for Cancer-Associated Fibroblasts Differentiation and Invasion In Vitro Studies

Published: August 8, 2019 doi: 10.3791/60122

Summary

The goal of this protocol is to establish a 3D in vitro model to study the differentiation of cancer-associated fibroblasts (CAFs) in a tumor bulk-like environment, which can be addressed in different analysis systems, such as immunofluorescence, transcriptional analysis and life cell imaging.

Abstract

Defining the ideal model for an in vitro study is essential, mainly if studying physiological processes such as differentiation of cells. In the tumor stroma, host fibroblasts are stimulated by cancer cells to differentiate. Thus, they acquire a phenotype that contributes to the tumor microenvironment and supports tumor progression. By using the spheroid model, we have set up such a 3D in vitro model system, in which we analyzed the role of laminin-332 and its receptor integrin α3β1 in this differentiation process. This spheroid model system not only reproduces the tumor microenvironment conditions in a more accurate way, but also is a very versatile model since it allows different downstream studies, such as immunofluorescent staining of both intra- and extracellular markers, as well as deposited extracellular matrix proteins. Moreover, transcriptional analyses by qPCR, flow cytometry and cellular invasion can be studied with this model. Here, we describe a protocol of a spheroid model to assess the role of CAFs' integrin α3β1 and its ectopically deposited ligand, laminin-332, in differentiation and in supporting the invasion of pancreatic cancer cells.

Introduction

The tumor microenvironment is a very complex niche and extremely important for the maintenance and progression of the tumor cells1. It is formed not only by the cancer cells but also by stromal fibroblasts. The tumor cells are surrounded by a stroma that is specific and different from the stroma of normal tissues2. Laminin-332 is an extracellular matrix protein ectopically expressed in the stroma of different tumors, such as of pancreatic adenocarcinoma3. Moreover, the biochemical composition of the ECM and also its biophysical properties, such as rigidity and tension, change within the tumor bulk4. This tumor stroma, or "reactive stroma", is caused by an adaptation of fibroblasts to the neighboring cancer cells and by the recruitment of other very important players that develop a favorable and supportive environment for tumor progression. The differentiation of stromal fibroblasts results in cancer-associated fibroblasts (CAF). These cells can be identified using different markers such as α-smooth muscle actin (αSMA)5 or neural/glial antigen 2 (NG2)6.

The most suitable in vitro model to recapitulate the tumor microenvironment (TME) with CAFs is difficult to select. The method to mimic physiological parameters of the TME in a cost-efficient and reproducible way must be considered for such a model system. Within the TME, different processes, such as proliferation, differentiation, migration and invasion of the different cell types occur. These cellular processes can be performed individually with different methods. However, the experimental conditions must consider the cellular interactions with the tumor stroma ECM, since the stiffness of the substratum influences the CAF differentiation process. R.G. Wells commented on the impact of matrix stiffness on cell behavior and highlighted that cytoskeletal organization and differentiation status observed in in vitro cultured cells might be artefactual7. Different stimuli seem to be involved in CAF differentiation, including mechanical tension5,7. To avoid this, 2D soft substrates could be possible approaches for differentiation studies, as they circumvent the problem of the stiff culture dish plastic. A soft 2D surface, on which fibroblasts can be grown, can be collagen-I coated polyacrylamide gels, whereby the gel stiffness can be manipulated by the concentration of polyacrylamide and the gel cross-linker. The adhesion and formation of αSMA-rich stress fibers are enhanced in fibroblasts along with the gel stiffness8. These results stress the importance of soft substrate scaffolds for more physiological in vitro differentiation models. However, in our hands the experimental reproducibility and imaging of these gels were challenging. To overcome these shortcomings, we changed the 2D soft substrate system for a 3D spheroid model for differentiation and invasion studies. This model is more clinically relevant and, similar to an in vitro organoid, recapitulates in vivo cell-cell interactions, ECM production and deposition, as well as cell behavior9.

Spheroids are formed when cells lack a substrate to adhere to. When the cells are left without an adhesive surface, they aggregate to form a more or less spherical structure. If the spheroids are composed of one type of cell, they are called homospheroids; if composed of two or more different cell types they form heterospheroids.

Among the different methods for spheroid preparation, we perform the protocol using non-adherent round bottom 96-well plates. It is very effective with respect to the costs. Here, we produce both homospheroids of fibroblasts, CAF or CAFs lacking the integrin α3 subunit to examine the differentiation process and heterospheroids of CAFs or integrin α3 KO CAFs and pancreatic duct carcinoma cells (AsPC-I and PANC-I) to study the invasion into the surrounding matrix.

The aim for these studies was to use primary CAFs isolated from human pancreatic carcinoma biopsies. However, the biopsies to obtain the cells are scarce and for this reason, the CAFs used in these studies have been immortalized using lentivirus containing HTERT. They are called iCAFs, and their normal counterparts, primary human pancreatic fibroblasts, are termed iNFs. The human pancreatic fibroblasts and the pancreatic duct carcinoma cells, AsPC-I and PANC-I, are commercially available.

This protocol was used to study the effect of the laminin-332-integrin interaction in the CAF differentiation process. To prove specificity of this interaction and its function, inhibitor compounds were used: BM2, a monoclonal antibody that blocks the integrin binding site the laminin-332 α3 chain10, or lebein 1, a snake venom derived compound that blocks the laminin-binding integrins α3β1, α6β1 and α7β111,12.

For the invasion assay, cells had been transduced with lentivirus containing cDNA encoding either mCherry (iCAFs and integrin α3 KO iCAFs) or GFP (AsPC-I and PANC-I) to distinguish the different cell types in the heterospheroids. The transduction of the cells to immortalize them and/or to label them with fluorescent protein (mCherry and GFP) expression is described in a previous study13, that should be consulted for further information.

Subscription Required. Please recommend JoVE to your librarian.

Protocol

1. 3D spheroids as an in vitro model for fibroblast/CAF differentiation using different TGF-β1 inhibiting compounds of cell-matrix interaction

  1. Mix 1 part of 6 mg/mL methylcellulose solution and 3 parts of cell culture media, MEM supplemented with 1% of heat-inactivated fetal bovine serum (FBS) and 1% penicillin/streptomycin to obtain the spheroid formation solution.
  2. Resuspend freshly thawed immortalized normal fibroblasts (iNFs), immortalized CAF (iCAFs) or integrin α3β1 KO iCAF (passage up to 25) in the spheroid formation solution. If preparing one 96 well plate, prepare an excess of spheroid formation solution, 10 mL and add 75,000 cells, keeping in mind that each spheroid is composed by 750 cells, and that 100 µL are distributed per each well of the plate.
  3. If cytokines or integrin inhibitors are included in the study, add these compounds to the cell suspension, in order to embed them into the spheroids during their formation. Note that iNFs are stimulated with 10 ng/mL of TGF-β1 in order to trigger differentiation. Where appropriate, add inhibitor compounds together with TGF-β1, such as 20 µg/mL BM2 or 10 µg/mL of lebein 1.
  4. Prepare CAF homospheroids in the same way but without the TGF-β1 stimulus, since they are already differentiated. Prepare the integrin α3 KO CAF homospheroids without the addition of any compound.
  5. Distribute the spheroid formation solution onto a round bottom 96 multi-well plate by adding 100 µL of the solution in each well. Every time the spheroid solution is distributed in the wells, mix well by pipetting up and down, several times.
  6. Place the plate in the cell culture incubator for 24 h to allow one spheroid to be formed in each well.
  7. Collect the spheroids after about 24 h and use them for different downstream purposes: immunofluorescent staining, real time (RT) q-PCR and flow cytometry. To detect expression of intra- and extracellular antigens, including deposition of ECM proteins within the spheroid, use immunofluorescent staining. After disintegration of spheroids into single cells, quantify membrane-anchored receptor by flow cytometry. To detect gene activation and transcriptional changes, use RT q-PCR of mRNA isolated from the spheroid cells.

2. Immunofluorescent staining of spheroids

  1. Collect the spheroids after about 24 h into one 1.5 mL reaction tube per experimental condition or per protein to be analyzed. For example, place the spheroids of iNFs stimulated with TGF-β1 in a tube different from the control iNFs, which were not treated. To avoid rupture of spheroids due to too strong shear forces, cut the end of the pipette tip to enlarge the orifice. Include at least 5-10 spheroids in one reaction tube, to allow replicates and take into account the possible losses during the washing steps.
  2. Centrifuge the spheroids with a bench top centrifuge for about 30-60 s at 1,000 x g. Carefully remove the methylcellulose-containing supernatant by pipetting, without disturbing the pelleted spheroids.
  3. Wash with 50 µL of 1x PBS. Repeat the centrifugation step and carefully remove the PBS by pipetting, as described in 2.2.
  4. Depending on the protein to be stained, fix with 50 µL of 4% paraformaldehyde in PBS for 20 minutes at room temperature (for αSMA, NG2 and integrin α3β1) or with 50 µL of methanol for 10 min at -20 °C (for laminin-332 subunits).
  5. Repeat the washing step as explained in steps 2.2-2.3.
  6. Permeabilize the cells with 0.1% Triton-X in PBS for 4 min at room temperature.
  7. Repeat the washing step as explained in steps 2.2-2.3 three times.
  8. Incubate the spheroids in PBS containing 5% FBS and 2% BSA, for 1 h at RT to block non-specific protein interaction sites.
  9. Incubate the spheroids with 30 µL of primary antibodies in PBS, 2.5% FBS and 1% BSA at 4 °C overnight. The following primary antibodies were used: anti-αSMA Cy-3 conjugated and anti-NG2 at 5 µg/mL; BM2 anti-laminin α3, 6F12 anti-laminin β3 and anti-laminin γ2 20 µg/mL.
  10. Repeat the washing step as explained in steps 2.2-2.3 three times.
  11. Incubate the spheroids with 30 µL of secondary antibodies in PBS, 2.5% FBS and 1% BSA at RT for 90 min. The following secondary antibodies used were: Alexa 488 anti-rabbit and anti-mouse (5 µg/mL).
  12. Repeat the washing step as explained in steps 2.2-2.3 three times.
  13. Incubate the cells with 30 µL of DAPI staining solution for 4 min at room temperature.
  14. Repeat the washing step as explained in steps 2.2-2.3.
  15. Place the spheroids on a glass slide, in a drop of PBS, using a glass Pasteur pipet.
  16. Image the spheroids by fluorescence microscopy in a laser scanning microscope using a magnification of 10x. Take different optical cross-section of the spheroid at different optical planes of a z-stack (15-20 stack planes). To establish the laser settings in the microscope, use a spheroid composed of cells negative for the antigen of interest or a spheroid stained only with the secondary antibody. Reuse the same settings for all the samples that will be compared.
  17. Analyze the microscopic images with ImageJ software as published previously14. The steps are summarized in Supplemental Figure 1. As background, determine the integrated signal density of a cell-free region of interest (ROI). Calculate the total corrected cell fluorescence (TCCF) as:
    Equation 1
  18. Determine the total corrected fluorescence (TCF) of the spheroids as the TCCF normalized to the area of spheroid and the number of stacks.
    Equation 2

3. RT q-PCR of homospheroids

  1. To perform RT-qPCR, collect at least 288 spheroids (corresponding to three 96-well plates) into a 50 mL reaction tube. Centrifuge for 5 min at 1,000 x g and carefully remove the methylcellulose-containing supernatant by pipetting without disturbing the pelleted spheroids.
  2. Wash with 1 mL of 1x PBS and transfer the spheroids to a 1.5 mL reaction tube. Centrifuge the spheroids with a bench top centrifuge for about 30-60 s at 1,000 x g. Carefully remove the PBS by pipetting, without disturbing the pelleted spheroids.
  3. Dissociate the spheroids with 250 µL of 4 mg/mL collagenase B, 50 µg/mL DNase I, 2% BSA, 1 mM CaCl2 in PBS at 37 °C for about 30-45 min by gently vortexing and pulsing intermittently for a few seconds every 5 min.
  4. Repeat the washing step as explained in step 3.2.
  5. Isolate the total RNA from the cells of disintegrated spheroids using a commercial RNA isolation kit, according to the manufacturers’ instructions.
  6. Reverse transcribe the isolated mRNA into cDNA with a commercial kit, according to manufacturers’ instructions.
  7. Quantify the transcription levels in replicates by real-time PCR. The primers used were: α-SMA: Fw 5′-CCGACCGAATGCAGAAG GA-3′, Rev 5′ ACAGAGTATTTGCGCTCCGAA-3′15; Laminin α3 chain: Fw 5′-GCTCAGCTGTTTGTGGTTGA-3′, Rev 5′-TGTCTGCATCTGCCAATAGC-3′16; Laminin β3 chain: Fw 5′-GGCAGATGATTAGGGCAGCCGAGGAA-3′ Rev 5′-CGGACCTGCTGGATTAGGAGCCGTGT-3′17; Laminin γ2 chain: Fw 5′-GATGGCATTCACTGCGAGAAG-3′ Rev 5′-TCGAGCACTAAGAGAACCTTTGG-3′ 16; NG2: Fw 5′-CTGCAGGTCTATGTCGGTCA-3′ Rev 5′-TTGGCTTTGACCCTGACTATG-3′18; integrin α3 subunit: Fw 5’-AGGGGACCTTCAGGTGCA-3’ Rev 5’-TGTAGCCGGTGATTTACCAT-3’19; TOP-1: Fw 5′-CCAGACGGAAGCTCGGAAAC-3′ Rev 5′-GTCCAGGAGGCTCTATCTTGAA-3′20.
  8. Correct the Ct values for the Ct-value of an endogenous reference gene such as TOP-1 using the equation: 2- ΔΔCt. ΔΔCt = ΔCt (target sample) – ΔCt (reference sample) and ΔCt= Ct of target gene or reference gene – TOP-I.

4. Flow cytometry analysis of integrin expression

  1. For flow cytometric analysis, collect at least 288 spheroids (corresponding to three 96-well plates) into a 50 mL reaction tube. Centrifuge for 5 min at 1,000 x g and carefully remove the methylcellulose-containing supernatant by pipetting.
  2. Wash with 1 mL of 1x PBS and transfer the spheroids to a 1.5 mL reaction tube. Repeat the centrifugation step and carefully remove the PBS by pipetting.
  3. Dissociate the spheroids as described in step 3.3.
  4. Repeat the washing step as explained in step 4.2.
  5. To block non-specific binding sites and to prevent binding of detection-relevant antibodies to Fcγ-receptors (CD16, CD32, and CD64), incubate the cells in 100 µL of PBS containing 2% horse serum (or serum of another animal species, the antibodies of which will not be used in the experiment) and 0.1% BSA for 20 min at 4 °C with rotation.
  6. In case of staining intracellular markers, fix/permeabilize the cells as described under step 2.2-2.5.
  7. Incubate the cells with the primary antibodies in 100 µL of PBS containing 2% horse serum and 0.1% BSA for 30 min at 4 °C with rotation. Dilute the primary antibody to 10 µg/mL.
  8. Wash with 100 µL of PBS + 0.1% BSA three times, with centrifugation steps in the top bench centrifuge at 1,000 x g in between.
  9. Incubate the cells with the secondary antibodies in 100 µL of PBS containing 2% horse serum and 0.1% BSA for 30 min at 4 °C with rotation. Dilute secondary antibody to 10 µg/mL.
  10. Repeat the washing step as explained in step 4.7.
  11. Analyze 2.0 x 104 stained cells in a flow cytometer. Gate for single live cells via the FSC-SSC dot plot and analyze the fluorescence of the gated cells on the channel appropriate for the selected fluorophore.

5. Invasion assay using heterospheroids

  1. Prepare the spheroid formation solution for heterospheroids, containing the different cell types and corresponding mediums, as summarized in Table 1. The cells had previously been transduced with lentivirus containing vectors for either mCherry or GFP expression.
  2. Fill 100 µL of the spheroid formation solution in each well of the plate and incubate the plate in the cell culture incubator for 24 h to allow one spheroid to be formed in each well.
  3. Prepare 60 µL of gelatinous matrix mixture containing 2 mg/mL of rat tail collagen I, 30% of commercial gelifying matrix and 2.5 µg/mL of laminin-332 in a 1.5 mL reaction tube. Rapidly distribute 15 µL in 3 wells of the µ-slide angiogenesis chamber. Embed one spheroid in each well already containing the gel.
  4. If necessary, rapidly adjust the location of the spheroid to the center of the well with a pipette under the microscope.
  5. Repeat steps 5.2-5.3 for the next heterospheroids and repeat again for the homospheroids.
  6. Incubate the gels in the incubator for 1 h to solidify and, then, gently overlay the gels with cell culture medium.
  7. Image the spheroids by fluorescence microscopy in a laser scanning microscope. Take different optical cross-section of the spheroid at different optical planes of a z-stack and at different time points of invasion (e.g., 0 h, 24 h, 48 h and 72 h).
  8. Analyze the images by counting the number of invading cancer cells. Invading cells are those ones that have left the spheroid and entered the invasive area. The invasive area is defined as the ring area between the outer and inner perimeters given by the furthest invaded cell and by the spheroid border of the spheroid at the beginning of the invasion experiments, respectively, as outlined in Supplemental Figure 2.

Subscription Required. Please recommend JoVE to your librarian.

Representative Results

The results of this experimental design are published in Martins Cavaco et al.13, which is recommended for further reading on the conclusions that were drawn from these experiments.

Figure 1, a representative image of an immunofluorescent spheroid, shows the immunostaining of the integrin α3 subunit of both immortalized normal fibroblasts and immortalized CAFs (Figure 1A), as well as the immunofluorescence quantification (Figure 1B) and the transcriptional levels of the integrin α3 subunit gene by qPCR (Figure 1C). This panel of results demonstrates that integrin α3 is up-regulated in iCAFs, as compared to the normal counterpart. This proved that integrin α3β1 can be considered a marker for pancreatic fibroblasts differentiation. This was also demonstrated by flow cytometry studies13.

Figure 1
Figure 1. Expression of integrin α3 subunit by iNFs and iCAFs. The integrin α3 subunit is upregulated by iCAFs, reflecting its potential as a differentiation marker for pancreatic CAFs. (A) Immunofluorescent staining of homospheroids of normal pancreatic fibroblasts (iNFS) compared to the homospheroids of iCAF shows an increased signal of the integrin α3 subunit in the differentiated cells. (B) The fluorescence signal of the cells in the spheroid was quantified with the Z-stack images of the 3D spheroid, using the ImageJ software. Means ± SEM of three independent experiments are shown and compared by t-test (*, p < 0.1). (C) The cells obtained from the dissociated spheroids were analyzed for their transcriptional levels of integrin α3 subunit. Means ± SEM of ΔΔCt-values as fold changes from two independent experiments are shown and compared by t-test. Although not reaching significance level of 0.1, the transcriptional levels of integrin α3-encoding mRNA are higher in iCAFs than in iNFs. Please click here to view a larger version of this figure.

Heterospheroids
Cells type (passage up to 25) Number of cells Medium (1 part methylcellulose +…)
mCherry-iCAFs + GFP-AsPC-I 400 CAF + 400 pancreatic cancer cells 1,5 parts cell MEM with 10% of FBS and 1% pen/strep + 1,5 parts of RPMI with 10% of FBS and 1% pen/strep
mCherry-α3KO-iCAFs + GFP-AsPC-I
mCherry-iCAFs + GFP-PANC-I 1,5 parts cell MEM with 10% of FBS and 1% pen/strep + 1,5 parts of DMEM with 10% of FBS and 1% pen/strep
mCherry-α3KO-iCAFs + GFP-PANC-I
Homospheroids
Cell type (passage up to 25) Number of cells Medium (1 part methylcellulose +…)
mCherry-iCAFs 400 cells 3 parts MEM supplemented with 10% of FBS and 1% pen/strep
mCherry-α3KO-iCAFs
GFP-AsPC-I 3 parts RPMI with 10% of FBS and 1% pen/strep
GFP-PANC-I 3 parts DMEM with 10% of FBS and 1% pen/strep

Table 1. Cell composition of hetero- and homospheroids. The number of cells necessary to assemble the hetero- and homospheroids as well as the corresponding medium which should be used for the spheroid formation solution are summarized in the following table.

Supplemental Figure 1. Sequential steps for quantification of the immunofluorescent staining of a protein of interest in spheroids, using ImageJ. Please click here to download this file.

Supplemental Figure 2. Sequential steps for quantification of the number of invading cancer cells in the invasion assay, using ImageJ. Please click here to download this file.

Subscription Required. Please recommend JoVE to your librarian.

Discussion

To develop an appropriate in vitro model to study CAF differentiation is a challenging task. After employing different approaches, we concluded that a 3D spheroid model is the more practical, physiological and clinically relevant model, in which the interplay between pancreatic carcinoma cells with immortalized CAFs can be studied. This model prevented spontaneous differentiation of fibroblasts, due to artefactual stressors such as stiffness of the cell culture plastic, at least in short-term culture conditions (up to 48 h). Although the exact stiffness of the spheroid used in these studies was not measured, Ito et al. evaluated the stiffness of human umbilical vein endothelial cells and mesenchymal stem cell heterospheroids using a robot integrated microfluidic chip22. The average stiffness-index of the heterospheroids was measured 1 and 3 days after assembly. The size of the spheroids was different, the MSC seemed to increase their aggregation capacity, which resulted in a decrease in size over time (from 135.5 µm to 99.8 µm)21,22. Consequently, the stiffness of the spheroid also increased from 5.0 x 10-3 to 7.5 x 10-3 Pa22. However, the stiffness of the spheroids composed of a heterotypic population of CAFs and AsPC-I might possess mechanical properties and stiffness values different from the ones of homospheroids made of a single population of fibroblasts/CAFs. This is still unknown and might be different from what was described by Ito et al22. The stiffness also depends on the amount and cross-linkage of cell-produced and deposited ECM, on the different cell-cell interactions within the spheroid, on the number of cells within the spheroid or on the duration of culture.

Like any other model, the experimental design involves some variables that need optimization such as the number of cells necessary to form the spheroids, the time points of treatment and microscopic imaging. The described experimental design was optimized with the aim of having a spheroid size, which would have minimal influence in the diffusion of nutrients and oxygen, as there is a diffusion limit due to mass transport limitations9. For example, the transport of oxygen in spheroids with a size greater than 150-200 µm is significantly affected, as well as the diffusion of glucose and lactate, in aggregates bigger than 300 µm9,18. Spheroids with diameters larger than 400-500 µm usually present a concentrically layered structure consisting of a necrotic core surrounded by a viable layer of quiescent cells and an outer rim of proliferating cells9,24. To avoid the limited diffusion of compounds, and to eliminate the problem of the inaccessible spheroid core, cells were treated with TGF-β1, BM2 and lebein-1 prior to spheroid formation, when the cells are individually suspended in the spheroid formation solution. Moreover, to allow the availability of oxygen and nutrients to most of the cells in the spheroid and to prevent the formation of a necrotic core, the number of cells per spheroid was reduced to 750-1000 cells, which form spheroids with a diameter of 140-200 µm.

It is important to thoroughly mix the different cell types in the spheroid formation solution, so the spheroids are composed by approximately the same number of cells, avoiding significant size differences between the spheroids. Nevertheless, sometimes some spheroids can be slightly bigger than the other and that will result in more Z-stacks acquired in the microscope. Some spheroids might also deviate from the perfectly spherical form, but this is also taken in account when the ROI of the spheroid is delineated. These differences of size and shape are accounted in the normalized quantification values as explained in the next paragraph. Another important factor regarding the shape of the spheroid is the cell type. For example, fibroblasts and CAFs form spheroids with a nearly spherical form, while AsPC-I and PANC-I cells form a more disperse aggregate of cells.

For imaging the spheroids, cryosections of fixed spheroids can also be used, however the integrity of the spheroid may be compromised upon sectioning, depending on the cell type and the immunostaining protocol. Alternatively, the staining of the whole spheroid in its 3D structure, proved to be a simpler option. Acquiring microscopic Z-stacked pictures of the spheroid as a 3D structure takes on average 5-15 min per spheroid.

The immunofluorescent signals in images of spheroids are difficult to quantify as they represent planes of 3D structures. The method used was based on a previously described one, where the authors extrapolated and considered the spheroid to be a cell14. In this way, the fluorescence signal is corrected using Equation 1.

As background, the integrated signal density of a cell-free region of interest (ROI) was determined. The measured integrated signal density is the sum of the intensity values of the pixels within the selected ROI. Since spheroids are 3D structures, confocal images from different stacks are acquired. To process such images one can measure the fluorescent signal in each stack individually or use the sum of all stacks into a Z-projection. All the quantifications can be performed using ImageJ. The total corrected fluorescence (TCF) of the spheroids is determined as the normalized TCCF, considering the area of spheroid and the number of stacks, using Equation 2. This accounts for the varying sizes of spheroids and the discrepancy in the spherical shape. Analyzing the immunofluorescent staining of the spheroids using this method can take up to 5 min per spheroid.

Similarly, invasion of cells from spheroids into the surrounding stromal matrix can be analyzed by different algorithms. A straightforward method was previously described by Nowicki et al, in which the invasive cancer cells were counted23. In order to discriminate the invasive cells from the ones that do not invade, it is important to establish a spatial limit beyond which the cells are considered to have left the spheroid structure. Therefore, the starting point is the limit that corresponds to the perimeter of the spheroid images acquired at the initial time point (time 0), when the spheroids were embedded into the gel. The cell that invades the gel the furthest is considered the maximal distance an invasive cell can reach, and thus, it defines the outer rim of the invasion region. This method counts the invasive cancer cells that are present in the invading area, using the counting tool from ImageJ, and the ROI corresponding to the spheroid at time 0 h is added to the ROI manager and then transposed to the image of the same exact spheroid acquired 48 h or 72 h later. For this reason, it is important that the spheroid remains as close to the center of the planes as possible, during the different times of acquisition. Counting the number of cells can take up to 5 min per spheroid.

Another important consideration is to distinguish the different cell types in the heterospheroid. This can be performed using live cell fluorescent dyes, which can be problematic when the cell type has a high cell division rate or if the experiment is to be performed for long periods of time. The more robust and reliable way to distinguish the two different populations is to develop cell lines expressing fluorescent proteins such as mCherry and GFP. The delivery of the expression vectors can be performed using lentivirus, which results in stable expression of these proteins.

Subscription Required. Please recommend JoVE to your librarian.

Disclosures

The authors declare no conflict of interest. This material reflects only the author's views and the European Union is not liable for any use that may be made of the information contained therein.

Acknowledgments

We acknowledge Barbara Schedding's help in preparing the BM2 and lebein-1. We acknowledge Àgnes Noel for sharing her expertise in spheroid assays. We thank Sonja Schelhaas and Michael Schäfers for their help in handling lentiviral transfection under S2 conditions. We acknowledge Sabine von Rüden's assistence in preparing CAFs from pancreatic cancer tissue.

The research leading to these results has received funding from the People Programme (Marie Curie Actions) of the European Union's Seventh Framework Programme FP7/2007-2013/ under the REA grant agreement n◦ (316610) to J.A.E. Moreover, J.A.E. and A.C.M.C. was financially supported by the Deutsche Forschungsgemeinschaft (DFG) within the Cells-in-Motion Cluster of Excellence (EXC 1003-CiM). This project was also supported by Wilhelm Sander Stiftung (grant: 2016.113.1 to J.A.E.).

Materials

Name Company Catalog Number Comments
4',6-Diamidino-2-Phenylindole, Dihydrochloride (DAPI) SIGMA-ALDRICH D9542-10
6F12 anti-Laminin β3 subunit Mouse (monoclonal) Homemade
A3IIF5 Anti-α3 integrin subunit Mouse (monoclonal) Kindly provided by Prof. M. Hemler, Dana Faber-Cancer Institute, Boston
Acetone SIGMA-ALDRICH 32201
Albumin Fraction V - BSA AppliChem A1391
Alexa fluor 488 Goat (polyclonal) anti-Mouse Invitrogen A11029
Alexa fluor 488 Goat (polyclonal) Rabbit Invitrogen A11034
Anti-laminin γ2 subunit Mouse (monoclonal) Santa Cruz sc-28330
Anti-NG2 Rabbit (polyclonal) Millipore, AB5320 Millipore AB5320
Anti-α-SMA-Cy3 Mouse (monoclonal) SIGMA-ALDRICH C6198
AsPC-1 cell line ATCC Kindly given by prof. Jorg Haier's Lab
Bench centrifuge Fisher Scientific 50-589-620 Sprout
BM2 anti-laminin α3 subunit Mouse (monoclonal) Kindly provided by Prof. Patricia Rousselle, CNRS, Lyon
Calcium Chloride (CaCl2) Fluka 21074
Centrifuge Thermo Scientific Multifuge 1S-R
Centrifuge tubes 50 mL Corning 430290
Collagenase B Roche 11088831001
Collagen-I, rat tail Gibco A10483-01
Confocal microscope Zeiss LSM 700 and 800
DMEM (High glucose 4.5 g/L) Lonza BE12-604F
Dnase I Roche 10104159001
Flow Cytometer BD Biosciences FACSCaliburTM
Gelifying matrix ThermoFisher Scientific A1413202 Matrigel, Geltrex
Goat IgG, isotype DAKO X 0907
Horse Serum SIGMA-ALDRICH 12449-C
Human Primary Pancreatic Fibroblasts PELOBiotech PB-H-6201
Incubator Heraeus B6060
Laminin-332 Biolamina LN332
MEM SIGMA-ALDRICH M4655
Microplate, 96 wells, U-bottom Greiner Bio-One 650101
Microscope Slides Thermo Scientific J1800AMNZ
Mouse IgG, isotype SIGMA-ALDRICH I8765
Multi axle rotating mixer CAT RM5 80V
PANC-I ATCC Kindly given by Prof. Jorg Haier's Lab
Paraformaldehyde Riedel-de Haën 16005
Penicillin/streptomycin Gibco 15140-122
QuantiTect Reverse Transcription Kit Qiagen 205310
Rat IgG, isotype Invitrogen 10700
Reaction tubes, 1.5 mL Greiner Bio-One 616201
Real-time PCR cycler Qiagen Rotor-Gene Q
RNeasy Mini Kit Qiagen 74104
Rotor Gene SYBR Green PCR Kit Qiagen 204074
RPMI Lonza BE12-702F Add glucose to 4.5 g (0.2 um filter) and 1% sodium pyruvate
TritonX-100 SIGMA-ALDRICH X100RS
Vórtex Scientific Industries Vortex-Genie 2
μ-Slide Angiogenesis, uncoated Ibidi 81501

DOWNLOAD MATERIALS LIST

References

  1. Pietilä, M., Ivaska, J., Mani, S. A. Whom to blame for metastasis, the epithelial-mesenchymal transition or the tumor microenvironment? Cancer Letters. 380 (1), 359-368 (2016).
  2. Heldin, C. -H., Rubin, K., Pietras, K., Ostman, A. High interstitial fluid pressure - an obstacle in cancer therapy. Nature Reviews. Cancer. 4 (10), 806-813 (2004).
  3. Tani, T., et al. Pancreatic carcinomas deposit laminin-5, preferably adhere to laminin-5, and migrate on the newly deposited basement membrane. The American Journal of Pathology. 151 (5), 1289-1302 (1997).
  4. Eble, J. A., Niland, S. The extracellular matrix in tumor progression and metastasis. Clinical & Experimental Metastasis. , (2019).
  5. Tomasek, J. J., Gabbiani, G., Hinz, B., Chaponnier, C., Brown, R. A. Myofibroblasts and mechano-regulation of connective tissue remodelling. Nature Reviews. Molecular Cell Biology. 3 (5), 349-363 (2002).
  6. Sugimoto, H., Mundel, T. M., Kieran, M. W., Kalluri, R. Identification of fibroblast heterogeneity in the tumor microenvironment. Cancer Biology & Therapy. 5 (12), 1640-1646 (2006).
  7. Wells, R. G. The role of matrix stiffness in regulating cell behavior. Hepatology (Baltimore, Md.). 47 (4), 1394-1400 (2008).
  8. Cavaco, A., Rezaei, M., Niland, S., Eble, J. A. Collateral Damage Intended-Cancer-Associated Fibroblasts and Vasculature Are Potential Targets in Cancer Therapy. International Journal of Molecular Sciences. 18 (11), 2355 (2017).
  9. Mehta, G., Hsiao, A. Y., Ingram, M., Luker, G. D., Takayama, S. Opportunities and challenges for use of tumor spheroids as models to test drug delivery and efficacy. Journal of Controlled Release: Official Journal of the Controlled Release Society. 164 (2), 192-204 (2012).
  10. Rousselle, P., Lunstrum, G. P., Keene, D. R., Burgeson, R. E. Kalinin: an epithelium-specific basement membrane adhesion molecule that is a component of anchoring filaments. The Journal of Cell Biology. 114 (3), 567-576 (1991).
  11. Eble, J. A., Bruckner, P., Mayer, U. Vipera lebetina venom contains two disintegrins inhibiting laminin-binding beta1 integrins. The Journal of Biological Chemistry. 278 (29), 26488-26496 (2003).
  12. Kusuma, N., et al. Integrin-dependent response to laminin-511 regulates breast tumor cell invasion and metastasis. International Journal of Cancer. 130 (3), 555-566 (2012).
  13. Martins Cavaco,, C, A., et al. The Interaction between Laminin-332 and α3β1 Integrin Determines Differentiation and Maintenance of CAFs, and Supports Invasion of Pancreatic Duct Adenocarcinoma Cells. Cancers. 11 (1), 14 (2019).
  14. Ansari, N., Müller, S., Stelzer, E. H. K., Pampaloni, F. Quantitative 3D cell-based assay performed with cellular spheroids and fluorescence microscopy. Methods in Cell Biology. 113, 295-309 (2013).
  15. Goldberg, M. T., Han, Y. -P., Yan, C., Shaw, M. C., Garner, W. L. TNF-alpha suppresses alpha-smooth muscle actin expression in human dermal fibroblasts: an implication for abnormal wound healing. The Journal of Investigative Dermatology. 127 (11), 2645-2655 (2007).
  16. Kim, B. G., et al. Laminin-332-Rich Tumor Microenvironment for Tumor Invasion in the Interface Zone of Breast Cancer. The American Journal of Pathology. 178 (1), 373-381 (2011).
  17. Chen, J., et al. Overexpression of β3 Chains of Laminin-332 is Associated With Clinicopathologic Features and Decreased Survival in Patients With Pancreatic Adenocarcinoma. Applied Immunohistochemistry & Molecular Morphology. 23 (7), 516-521 (2015).
  18. Huang, C. -C., et al. Hypoxia-induced therapeutic neovascularization in a mouse model of an ischemic limb using cell aggregates composed of HUVECs and cbMSCs. Biomaterials. 34 (37), 9441-9450 (2013).
  19. Wang, L., Wang, L., Gu, Y., Shu, Y., Shen, Y., Xu, Q. Integrin α6high Cell Population Functions as an Initiator in Tumorigenesis and Relapse of Human Liposarcoma. Molecular Cancer Therapeutics. 10 (12), 2276-2286 (2011).
  20. Makizumi, R., Yang, W. -L., Owen, R. P., Sharma, R. R., Ravikumar, T. S. Alteration of Drug Sensitivity in Human Colon Cancer Cells after Exposure to Heat: Implications for Liver Metastasis Therapy using RFA and Chemotherapy. International Journal of Clinical and Experimental Medicine. 1 (2), 117-129 (2008).
  21. Saleh, F. A., Whyte, M., Genever, P. G. Effects of endothelial cells on human mesenchymal stem cell activity in a three-dimensional in vitro model. European Cells & Materials. 22, 242-257 (2011).
  22. Ito, K., Sakuma, S., Kimura, M., Takebe, T., Kaneko, M., Arai, F. Mechanical characterization system using on-chip probe with wide range actuation. 2016 International Symposium on Micro-NanoMechatronics and Human Science (MHS). , 1-2 (2016).
  23. Curcio, E., Salerno, S., Barbieri, G., De Bartolo, L., Drioli, E., Bader, A. Mass transfer and metabolic reactions in hepatocyte spheroids cultured in rotating wall gas-permeable membrane system. Biomaterials. 28 (36), 5487-5497 (2007).
  24. Lin, R. -Z., Lin, R. -Z., Chang, H. -Y. Recent advances in three-dimensional multicellular spheroid culture for biomedical research. Biotechnology Journal. 3 (9-10), 1172-1184 (2008).
  25. Nowicki, M. O., et al. Lithium inhibits invasion of glioma cells; possible involvement of glycogen synthase kinase-3. Neuro-Oncology. 10 (5), 690-699 (2008).

Tags

3D Spheroid Model Physiological System Cancer-associated Fibroblasts Differentiation Invasion In Vitro Studies Tumor Tissue Recapitulation 2D System Stiff Plastic Fibroblast Differentiation Cell Matrix Interaction Cell Culture Media MEM FBS Penicillin Streptomycin Methylcellulose Solution Spheroid Formation Solution Normal Fibroblasts Cytokines Integrin Inhibitors TGF Beta One
A 3D Spheroid Model as a More Physiological System for Cancer-Associated Fibroblasts Differentiation and Invasion In Vitro Studies
Play Video
PDF DOI DOWNLOAD MATERIALS LIST

Cite this Article

Cavaco, A. C. M., Eble, J. A. A 3DMore

Cavaco, A. C. M., Eble, J. A. A 3D Spheroid Model as a More Physiological System for Cancer-Associated Fibroblasts Differentiation and Invasion In Vitro Studies. J. Vis. Exp. (150), e60122, doi:10.3791/60122 (2019).

Less
Copy Citation Download Citation Reprints and Permissions
View Video

Get cutting-edge science videos from JoVE sent straight to your inbox every month.

Waiting X
Simple Hit Counter