Waiting
Login processing...

Trial ends in Request Full Access Tell Your Colleague About Jove

Cancer Research

Multidimensional Coculture System to Model Lung Squamous Carcinoma Progression

Published: March 17, 2020 doi: 10.3791/60644

Summary

An in vitro model system was developed to capture tissue architectural changes during lung squamous carcinoma (LUSC) progression in a 3-dimensional (3D) co-culture with cancer-associated fibroblasts (CAFs). This organoid system provides a unique platform to investigate the roles of diverse tumor cell-intrinsic and extrinsic changes that modulate the tumor phenotype.

Abstract

Tumor-stroma interactions play a critical role in the development of lung squamous carcinoma (LUSC). However, understanding how these dynamic interactions contribute to tissue architectural changes observed during tumorigenesis remains challenging due to the lack of appropriate models. In this protocol, we describe the generation of a 3D coculture model using a LUSC primary cell culture known as TUM622. TUM622 cells were established from a LUSC patient-derived xenograft (PDX) and have the unique property to form acinar-like structures when seeded in a basement membrane matrix. We demonstrate that TUM622 acini in 3D coculture recapitulate key features of tissue architecture during LUSC progression as well as the dynamic interactions between LUSC cells and components of the tumor microenvironment (TME), including the extracellular matrix (ECM) and cancer-associated fibroblasts (CAFs). We further adapt our principal 3D culturing protocol to demonstrate how this system could be utilized for various downstream analyses. Overall, this organoid model creates a biologically rich and adaptable platform that enables one to gain insight into the cell-intrinsic and extrinsic mechanisms that promote the disruption of epithelial architectures during carcinoma progression and will aid the search for new therapeutic targets and diagnostic markers.

Introduction

Lung cancer is the leading cause of cancer-related mortality worldwide. Lung squamous cell carcinoma (LUSC), which is the second most common type of non-small-cell lung cancer (NSCLC) and accounts for approximately 30% of all lung cancer, is often diagnosed at advanced stages and has a poor prognosis1. Treatment options for LUSC patients are a major unmet need that can be improved by a better understanding of the underlying cellular and molecular mechanisms that drive LUSC tumorigenesis.

As with most human cancers, the pathogenesis of LUSC is characterized by the disruption of the intact, well-ordered epithelial tissue architecture2. During this process, proper apical-basal cell polarity, cell-cell and cell-matrix contacts are lost, permitting uncontrolled growth and invasive behavior of the tumor cells. It is now widely appreciated that the malignant features of cancer cells cannot be manifested without an important interplay between cancer cells and their local tumor microenvironment (TME)3. Key components in the TME including extracellular matrix (ECM), cancer-associated fibroblasts (CAFs) as well as endothelial cells and infiltrating immune cells actively shape the TME and drives tumorigenesis4. Nevertheless, our current understanding of how the tumor cells and these key components in the TME interact to drive tissue architectural changes during LUSC progression is very limited.

Three-dimensional (3D) culture is an important tool to study the biological activities of cell-intrinsic and extrinsic changes in regulating tissue architectural changes in both normal and diseased tissues5. 3D cultures provide the appropriate structural and functional context that is usually lacking in traditional two-dimensional (2D) cultures. The added dimensions of such systems more closely mimic tissue in vivo in many aspects of cell physiology and cellular behaviors, including proliferation, differentiation, migration, protein expression and response to drug treatment. In recent years, efforts from various labs have led to the development of in vitro 3D models for both the normal lung as well as NSCLC6,7,8. However, a model for lung squamous carcinoma that can recapitulate both the dynamic tissue architectural changes during tumorigenesis as well as incorporate key stromal components was unavailable.

Here, we describe the methods for establishing a novel 3-dimensional (3D) coculture system using primary PDX-derived LUSC cells (termed TUM622) and CAFs9,10. Both TUM622 and CAFs are derived from NSCLC patient with poorly differentiated tumors10. When embedded as single cells in ECM, a rare subpopulation of TUM622 cells have the capacity to form organoids with acinar-like structures that display proper apical-basal cell polarity. These acinar-like structures are hyperplastic, display heterogeneous expression of stem-like and differentiation markers similar to the original tumor while remaining non-invasive, and thus mimic the earliest stage of LUSC development. Importantly, we showed that the tissue architecture of the acinar-like structures could be altered by inhibition of cell-intrinsic signaling pathways with small molecule inhibitors or addition of key components in the ECM such as CAFs, the latter of which enhances acini formation and further provokes the acini to become invasive when in close proximity. Together, these data suggest that this 3D co-culture system of LUSC organoids provides a valuable platform for the investigation of the dynamic reciprocity between LUSC cells and the TME and could be adapted for monitoring the response of LUSC cells to drug treatment11.

Subscription Required. Please recommend JoVE to your librarian.

Protocol

1. Passaging and Culturing TUM622 Cells and CAFs in 2D Cultures

  1. Passaging and culturing TUM622 cells
    1. Warm 3D culture medium and cell dissociation reagents (see Table of Materials) for TUM622 cells at 37 °C.
    2. Passage TUM622 cells at 80% confluency in 2D flasks. Usually, this occurs 1 week after passaging.
    3. Discard old medium from a T75 flask and wash once with 6 mL of HEPES buffer. Avoid pipetting directly onto the cells.
    4. Aspirate the HEPES buffer. Add 4 mL of trypsin/EDTA (0.25 mg/mL, see Table of Materials) for a quick rinse and discard the trypsin/EDTA.
    5. Add 2 mL of trypsin/EDTA and incubate at 37 °C for 5 min. Remove flasks from the incubator and tap the flasks to loosen the cells without creating air bubbles and return flasks to the incubator for an additional 5 min.
      NOTE: Prolonged exposure to trypsin will irreversibly damage the cells and alter their phenotype, thus it is recommended to limit the time cells are exposed to trypsin.
    6. Confirm cells have detached and dissociated under a light microscope (4x or 10x). Add 4 mL of neutralization buffer (TNS buffer) (see subculture reagent information in the Table of Materials) followed by 10 mL of 3D culture medium (see Table of Materials).
    7. Pipette up-and-down gently to further dissociate the cells using a 10 mL pipette. Transfer the suspension through a 40 µm cell strainer into a 50 mL conical tube.
    8. Count cell numbers using a hemocytometer or automated cell counter.
    9. Seed 0.8 x 106 cells/T75 flask in 20 mL of 3D culture medium (see Table of Materials).
    10. Feed the cells every other day by replacing half of the spent medium with fresh medium.
  2. Passaging and culturing CAFs
    1. Passage CAFs when cells reach confluency. Usually, this occurs after 5 days of culturing from a 1:2 split.
    2. Prepare CAF medium using RPMI basal medium with 20% heat-inactivated fetal bovine serum, 1% L-Glutamine and 1% Penicillin/Streptomycin. Warm the medium to 37 °C.
    3. In a T75 flask, rinse CAFs with phosphate-buffered saline (PBS) once then add 2 mL of trypsin/EDTA and incubate at 37 °C for 5 min.
    4. Observe under a light microscope to ensure cells have dissociated in the flask (4x or 10x). If not, extend the incubation for another 2-3 min.
    5. Once cells have detached and dissociated, add 10 mL of 3D culture medium to neutralize the trypsin/EDTA and pipette up and down several times to further dissociate the CAFs.
    6. Transfer the cell suspension into a 50 mL conical tube and spin down at 300 x g for 5 min at room temperature.
    7. Discard the supernatant and resuspend the pellet in an appropriate volume of 3D culture medium (see Table of Materials) and passage into two new T75 flasks.

2. Plating TUM622 Cells in the Extracellular Matrix for 3D Culturing

  1. The day before the experiment, thaw vials of basement membrane matrix in a 4 °C refrigerator overnight. Cooldown plastic pipettes (2 mL) and tips at -20 °C overnight.
    NOTE: Not all lots of basement membrane matrix have the same capacity to support the 3D growth of TUM622 cells. Therefore, it is necessary to acquire and test multiple lots of basement membrane matrix to identify those that support robust acini formation. Usually, this requires a higher protein concentration (16-18 mg/mL) in the matrix.
  2. On the day of the experiment, warm 3D culture medium, HEPES buffer, trypsin/EDTA and trypsin neutralization buffer (TNS) in a 37 °C water bath. Immediately before setting up the culture, take the thawed basement membrane matrix out of the fridge and put the vial on ice.
  3. Cooldown the tissue culture plates on a metal platform cooler placed on ice. Place centrifuge tubes on a metal cooling rack on ice.
  4. Using TUM622 cells obtained from step 1.1.7, calculate the desired number of cells needed for plating. Typically, 15,000-30,0000 cells are needed per well of a 24-well plate. Lower density is more suited for imaging and quantification, while higher density is preferred when collecting cells for RNA extraction or western blotting.
  5. Transfer cell suspension into a cooled centrifuge tube (each tube containing cells for triplicate plating) and spin down at 300 x g in a hanging bucket centrifuge at 4 °C for 5 min.
  6. Aspirate the supernatant carefully with an aspirating pipette attached to an unfiltered tip (20 µL), leaving approximately 100 µL of the medium in the tube (use markings on the tube as a guide).
  7. Gently tap on the side of the tube to dislodge and dissociate the pellet before returning it to the cooling rack.
  8. Using the 2 mL pre-cooled pipettes, gently mix the matrix by pipetting up and down a few times while keeping the vial in contact with the ice. Pipette at an even and moderate speed so that no bubbles are introduced into the matrix during this procedure.
  9. Transfer the appropriate volume of the matrix into each centrifuge tube. For plating triplicates in a 24-well plate, add 1.1 mL of basement membrane matrix to each tube.
  10. Using pre-cooled tips, pipette the matrix in each tube up and down about 10 times to make a uniform cell suspension.
  11. Transfer 310 µL of cell/matrix suspension into each well of a pre-cooled 24-well plate. The pipette is placed at a 90° angle to the plate surface and the suspension added to the center of the well. The suspension should spread and cover the entire well without needing to tilt the plate.
  12. To facilitate downstream immunofluorescence analysis, plate the cell/matrix suspension in parallel into 2-well chamber slides. Transfer 100 µL of cell/matrix suspension into the center of a well of 2-well chamber slide (see Table of Materials). This allows the matrix to form a dome-like structure with much smaller volume.
  13. Return the plate and the chamber slide back into a tissue culture incubator and incubate for 30 min to allow the matrix to solidify. Examine the plate/slide under a light microscope to ensure that single cells are evenly distributed within the matrix (4x or 10x).
  14. Add 1 mL of pre-warmed 3D culture complete medium into each well and 1.5 mL of 3D culture medium to each well of the chamber slide then return them to the incubator.

3. 3D Coculturing of TUM622 Cells and CAFs in the Extracellular Matrix

  1. Prepare cell suspensions of TUM622 and CAFs according to section 2.
  2. Count the CAF cell density by taking 10 µL of cell suspension and mixing it with 10 µL of trypan blue.
  3. Add 10 µL of the mixture to each of the two chambers on a hemacytometer to count and calculate cell density.
    NOTE: CAFs have irregular shapes and may not be accurately counted on an automatic cell counter.
  4. Co-embedding TUM622 cells and CAFs in basement membrane matrix
    1. Based on the cell density information, calculate the desired number of cells used for plating. CAFs are seeded at a 2:1 ratio of TUM622 cells. For example, for 30,000 TUM622 cells seeded, 60,000 CAFs are co-embedded.
    2. Transfer the appropriate volume of TUM622 as well as CAFs cell suspension into the same centrifuge tube and follow steps 2.5-2.11 for plating into 24-well plates. For immunofluorescence, transfer 60 µL of TUM622/CAFs mix to chamber slides as described in step 2.12).
  5. Coculturing TUM622 with overlaid CAFs in basement membrane matrix (see Table of Materials)
    1. Set up TUM622 mono-culture according to steps 2.5-2.13.
    2. Transfer twice the number of CAFs suspension (compared to the number of TUM622 cells seeded) into a centrifuge tube and spin down at 300 x g for 5 min at room temperature.
    3. Aspirate the supernatant and resuspend the CAFs in 1 mL of 3D culture medium.
    4. Transfer the 1 mL of CAFs suspension to the well containing the embedded TUM622 cells.

4. Harvesting TUM622 Acini for RNA/Protein Extraction and Fluorescence-activated Cell Sorting (FACS)

  1. Prepare wash buffer and cell harvesting buffer according to the 3D cell harvesting kit protocol the previous day and chill overnight at 4 °C.
  2. Keep plates on a plate cooler and other reagents on ice before starting the extraction process.
  3. Aspirate media from 3D culture wells without touching the matrix and gently wash the well 3 times with 1 mL of wash buffer.
  4. Aspirate the final wash and add 1 mL of cell harvesting buffer to each well.
  5. Use a p1000 pipette tip to scrape the matrix off of each well.
  6. Pipette up and down to further dissociate the matrix.
  7. Transfer 1 mL of the mix to a pre-chilled 15 mL conical tube. Add another 1 mL of harvesting buffer to the same well.
  8. Repeat steps 4.5-4.7, and transfer all mix of the same well into one 15 mL conical tube.
  9. Cap the tubes and rock at 4 °C for 30 min.
  10. Fill each tube with ice-cold PBS up to 10 mL and then centrifuge at 300 x g for 5 min at 4 °C.
  11. Aspirate the supernatant without touching the pellet. The supernatant should contain matrix fragments, but the spheroids should all be collected at the bottom of the tube.
  12. Add ice-cold PBS for a second wash. Invert the tube a few times to dissociate the pellet. Spin down at 300 x g for 5 min.
  13. While spinning, prepare lysis buffer for protein and RNA collection.
  14. Carefully aspirate the supernatant and add lysis buffer for downstream processing to collect protein or RNA. Alternatively, cells could be resuspended for flow analysis/FACS sorting or serial passaging.

5. Immunofluorescence of TUM622 Acini

  1. Prepare immunofluorescence buffer (IF buffer: PBS with 0.1% bovine serum albumin (BSA), 0.2% Triton X-100 and 0.05% Tween-20), primary blocking buffer (IF buffer with 10% goat serum), secondary blocking buffer (primary blocking buffer with 20 µg/mL goat anti-mouse F(ab')2)
  2. Aspirate medium from 2-well chamber slides, rinse once with PBS and set the slide on metal plate cooler on ice. The chamber slide should remain on the metal plate cooler for the remainder of the protocol.
  3. Add pre-chilled 4% PFA to fix the acini and incubate on ice for 20 min.
  4. Remove 4% PFA and wash three times with 2 mL of pre-chilled PBS each for 5 min with gentle rocking on a rocker.
  5. Aspirate PBS and permeabilize with 1.5 mL of 0.5% Triton X-100 in PBS (pre-chilled) for 20 min. By the end of this procedure, the dome-like structure will become loose.
  6. Gently aspirate the permeabilization buffer from the chamber slide to avoid sample loss. This is achieved by adding a fine tip (20 µL) to the aspirating pipette and pressing the tip towards the corner of the chamber.
  7. Wash three times with 2 mL of pre-chilled PBS each for 5 min with gentle rocking on a rocker.
  8. Block the sample with the primary blocking buffer on ice for 1 h.
  9. Remove primary blocking buffer and add secondary blocking buffer and block for 30 min.
  10. Add primary antibodies in primary blocking buffer and incubate overnight at 4 °C.
    NOTE: The concentration of the antibodies used here should be higher than normally used for staining cells in 2D culture. Most of the primary antibodies used in this study are diluted at 1:100 dilution (see Table of Materials).
  11. Remove primary antibodies and wash the sample 3 times with 2 mL of cold IF buffer.
    NOTE: The samples could be loose, take extra caution when aspirating.
  12. Incubate the samples in secondary antibodies diluted in primary blocking buffer for 1 h at RT. The preferred secondary antibodies should be highly cross-adsorbed to reduce background staining. Most secondary antibodies used in this study are diluted at 1:200 dilution.
  13. Remove secondary antibodies and wash the sample 3 times with 2 mL of cold IF buffer.
    NOTE: The samples could be loose, take extra caution when aspirating.
  14. Add PBS with DAPI (1:1,000 dilution) during the last wash to stain the nucleus. The perform another 2 washes in PBS.
  15. Image the samples on a confocal microscope within 3 days.
    NOTE: Due to the size of the organoids and limits in the objective's working distance, samples are usually imaged at 10x or 20x magnification.

6. Preparing 3D Culture Samples for Immunohistochemistry

  1. Aspirate medium from 2-well chamber slides and rinse once with PBS.
  2. Fix 3D cultures in 4% PFA at 37 °C overnight.
  3. Remove 4% PFA, surround cultures with 2.5 mL of histology sample gel (see Table of Materials) and place the slide at 4 °C to solidify for at least 1 h.
  4. Transfer samples surrounded with histological sample gel to tissue cassettes and processed in an automated tissue sample processor overnight.
  5. Embed samples in paraffin wax and prepare for sectioning12.

7. 3D Cytotoxicity Assay for Compound Screening (Example for One 96-well Plate)

  1. Set a 96-well plate on a plate cooler, a 25 mL reservoir on a reservoir cooler and a 15 mL conical tubes on ice before starting the experiment.
  2. Prepare cell matrix suspensions of TUM622 cells in a pre-chilled 15 mL polypropylene conical tube by adding the appropriate volume of basement membrane matrix to cells. The desired density for TUM622 cells is 10,000 cells per 70-75 µL of basement membrane matrix. Pipette up and down a few times to allow even mixing of cells within the matrix.
  3. Move the plate with plate cooler and reservoir with a reservoir cooler away from the ice to a dry surface to avoid contact of basement membrane matrix with ice during transfer.
  4. Transfer the matrix cell mixture to the cooling reservoir without creating bubbles.
  5. Using a mechanical multichannel pipette (10-300 µL), transfer 70-75 µL of the mix cells into each appropriate well of a 96-well plate.
  6. Incubate plate at 37 °C and 5% CO2 for 30 min for the basement membrane matrix to solidify.
  7. Add 100 µL of media in all rows and return the plate to the incubator.
  8. Start compound dosing the next day or later depending on the goal of the experiment.
  9. Spheroids can be re-fed and re-dosed every 2-3 days for up to 10 days, by removing spent media with an 8- or 12-well vacuum manifold and replacing with fresh media with or without desired compounds.
  10. The number of TUM622 spheroids could be quantified using 3D imager according to the manufacturer's protocol.

Subscription Required. Please recommend JoVE to your librarian.

Representative Results

TUM622 and CAFs in 2D culture
Figure 1 presents the typical morphology of TUM622 cells and CAFs in 2D culture. TUM622 cells are rounded with large nuclei while CAFs are flat and elongated. TUM622 cells can reach 80%-90% confluency in culture. Further proliferation leads to more, but smaller cells aggregated in colonies that do not come into direct contact. In contrast, CAFs prefer to grow at higher cell density and will keep proliferating at full confluency if sufficient nutrients are provided.

Growth and morphology of TUM622 acini in 3D ECM
Figure 2 presents a time-course experiment of TUM622 cells seeded in 3D culture. Data show that single TUM622 cells are capable of forming organoids with acinar-like morphologies when embedded. Between days 5 and 7, a lumen becomes apparent in the acinar-like structures and remains hollow thereafter (Figure 2A). Each acinus, composed of a monolayer of cells surrounding the hollow lumen, displays proper apical-basal polarity similar to that of lung epithelium in vivo (Figure 2B). These acinar-like structures are hyperplastic and continue to grow as long as sufficient nutrients are provided. The culture can be maintained for up to 24 days before the ECM completely disintegrates (Figure 2C). Through limiting dilution assay (LDA) (data not shown), it is estimated that only a rare subpopulation of TUM622 cells (<0.02%) have the capacity to form acinar-like structures9.

Growth and morphology of TUM622-CAFs coculture
Figure 3 depicts the setup and representative results of TUM622-CAF cocultures. CAFs could be integrated into the coculture by either overlaying on top of the matrix or co-embedded with the TUM622 cells. Regardless of the setup, the presence of CAFs greatly enhanced the number and size of the spheroids formed (Figure 3B). Interestingly, when TUM622 acini come into close proximity with CAFs, they induce the acini to become invasive and migrate towards the CAFs, forming "tear-drop" like structures (Figure 3C). Note that TUM622 acini in monoculture do not display invasive behavior and only form "tear-drop" like structures when close to CAFs.

Representative immunofluorescent and immunohistochemistry staining results
Figure 4 shows representative results from immunofluorescence and immunohistochemistry staining of TUM622 acini after 10 days of culturing. Confocal images were taken at the equatorial plane of immunofluorescently stained TUM622 acini (Figure 4A). In contrast, each section from the immunohistochemistry sample may capture acini at different planes (Figure 4B). Both results showed heterogeneous expression of stem-like and differentiated cells within each acinus.

TUM622 3D cytotoxicity assay using a Wnt pathway inhibitor
Figure 5 shows the dose-response of TUM622 acini treated with XAV939, a tankyrase inhibitor (Figure 5A,B). XAV939 was added to the culture 1 day after plating and refreshed every 2 days for a total of 10 days. At the end of the experiment, the number of acini was quantified by an imager. Brightfield images at higher magnification were also acquired to capture the morphology of spheroids in control versus XAV939-treated wells (Figure 5C). Overall, XAV939 displays dose-dependent inhibition on acini formation and alters the tissue architecture of the spheroids formed. These results suggest that activation of the canonical Wnt pathway is required during TUM622 acinar morphogenesis.

Figure 1
Figure 1: TUM622 and CAFs in 2D culture. Representative bright-field images of TUM622 cells and CAFs cultured in 2D. Scale bar = 100 µm. This figure has been modified from Chen et al.9 and used with permission. Please click here to view a larger version of this figure.

Figure 2
Figure 2: Growth and morphology of TUM622 acini in 3D ECM. (A) Time course images of TUM622 cells cultured in basement membrane matrix over a 10-day period. Scale bar = 100 µm. (B) Immunofluorescence of TUM622 acini stained with apical-basal cell polarity markers, Golgi-enzyme (GM-130, green, apical) and Integrin alpha 6 (CD49f, basal, red). (C) Quantification of acini number (right y-axis, red) and the average size of acini (left y-axis, blue) plated in triplicate in a 24-well plate over 24 days in culture. Error bars represent SD. This figure has been modified from Chen et al.9 and used with permission. Please click here to view a larger version of this figure.

Figure 3
Figure 3: Growth and morphology of TUM622-CAFs coculture. (A) Schematic drawing of the setup of TUM622-CAFs coculture. CAFs are overlaid or co-embedded with TUM62 cells in ECM. After 6-12 days in coculture, TUM622 cells are able to form more and larger acini compared with mono-culture and invade the ECM when in close proximity and direct contact with CAFs. Note that the invasive phenotype could only be observed in the co-culture. (B) Brightfield image of TUM622 3D cultures in the presence or absence of overlaid CAFs after 8 days. Scale bars = 200 µm. (C) Brightfield images showing tear-drop shaped acini forming in cocultures regardless of CAFs are overlaid or co-embedded in the ECM. Scale bars = 200 µm. This figure has been modified from Chen et al.9 and used with permission. Please click here to view a larger version of this figure.

Figure 4
Figure 4: Representative immunofluorescent and immunohistochemistry staining results. (A) Antibody staining of acini with markers of stem/progenitor cells (CXCR4 and SOX2), mesenchyme (Vimentin), epithelial differentiation (Involucrin), apoptosis (Cleaved-Caspase-3) and proliferation (Ki67) in green, DAPI in blue, E-cadherin and Phalloidin in red. Scale bar = 50 µm. (B) Immunohistochemistry on FFPE sections of TUM622 acini. Scale bar = 100 µm (top) and 50 µm (bottom). This figure has been modified from Chen et al.9 and used with permission. Please click here to view a larger version of this figure.

Figure 5
Figure 5: TUM622 3D cytotoxicity assay using a Wnt pathway inhibitor. (A) Quantification of spheroid numbers in a 96-well plate where TUM622 cells were treated with dimethyl sulfoxide (Control) or XAV939. Each condition is assayed in triplicates. Error bars represent SD. (B) Whole well images from a 24-well plate taken with an imager showing the inhibitory effects of XAV939 on acini formation. (C) Representative brightfield images from the control vs. treated wells demonstrating the morphological changes caused by XAV939 treatment. Scale bars = 100 µm. This figure has been modified from Chen et al.9 and used with permission. Please click here to view a larger version of this figure.

Subscription Required. Please recommend JoVE to your librarian.

Discussion

Tumors are heterogeneous tissues composed of cancer cells coexisting side-by-side with stromal cells such as cancer-associated fibroblasts, endothelial cells and immune cells within the ECM. Together, these diverse components cross-talk and influence the tumor microenvironment, playing an active role in driving tumorigenesis, a process that involves progressive changes in tumor architecture. Ideally, an in vitro model of tumor development should be able to capture the dynamic tissue architectural changes observed in human tumors in vivo, the complex interplay of diverse cell types within the tumor microenvironment and at the same time permit experimental manipulation on both the tumor cells and components in the TME. Although much progress has been made in 3D cancer models in recent years, such models have not been readily forthcoming for LUSC. Most models reported to date only incorporate a few aspects of these important features. Here we report the methods for a 3D coculture system of LUSC that simultaneously captures key tissue architectural changes observed during LUSC development as well as dynamic interactions between tumor cells and major components of the TME, including the ECM and CAFs.

The ability of this system to more accurately model tissue architectural changes is based on the unique property of TUM622 cells in forming organoids with acinar-like morphologies when embedded in 3D EC. Formed from a self-renewing single cell, each acinus is composed of a monolayer of cells surrounding a hollow lumen. This monolayer of cells exhibits apical-basal cell polarity and remains non-invasive, resembling the tissue architecture of the lung epithelium. While TUM622 as a 3D mono-culture display hyperplastic growth, the addition of CAFs further enhances acinar morphogenesis and induces more and larger acini to form. Importantly, CAFs invoke dynamic tissue architectural changes in TUM622 cells when the two cell types come into close proximity, allowing the TUM622 cells to lose their apical-basal polarity and invade the matrix toward the CAFs. These phenotypic changes recapitulate both early hyperplasia as well as late invasive stages of LUSC.

Unlike many tumor spheroid models where each spheroid is formed by aggregation of many cells, each TUM622 acinus is derived from a single cell9. By in vitro LDA, it is estimated that only a minor subpopulation (≤0.02%) of TUM622 cells have such capacity9. Although rare, these cells could self-renew as evidenced by their capacity to undergo serial passaging in 3D as well as differentiate into a heterogeneous population of cells similar to that of the original tumor. Due to this unique feature of TUM622 cells, it is critical to ensure even distribution of single TUM622 cells within the ECM at the time of plating for successful culturing and downstream analysis. To achieve this goal, several key points need to be followed carefully in the protocol, including the determination of appropriate seeding density, keeping all tools and reagents cool during the mixing of cells and matrix to prevent premature solidification, avoiding the introduction of bubbles during the mixing process and allowing sufficient time for matrix to fully solidify before adding culture medium. Together, these precautions will help to achieve a more uniform matrix substrate and culture condition for all embedded cells.

Once successfully established, this culture can be used for a variety of downstream analyses to dissect the cell and biochemical process that regulate tumorigenesis. The number and size of acini formed in each well can be monitored over time with bright field imagers and used as a readout for the proliferative and self-renewal capacity of TUM622 cells. More detailed dynamics in the morphogenesis of each acinus could be observed with live-imaging on a confocal microscope, with or without various labeling dyes. The conditioned medium can be collected at multiple time points during the culture period for analyzing soluble factors that may mediate cell-cell or cell-matrix cross-talks. TUM622 cells extracted directly from the ECM using protocol 4 are suitable for RNA and protein extraction for gene expression analysis, flow cytometry quantification or FACS sorting based on cell surface markers. Alternatively, the cultures could be fixed for in situ immunofluorescence or immunohistochemistry studies to understand the spatial-temporal distributions of various markers. Although similar, immunohistochemistry complements immunofluorescence methods in that it allows the sampling of entire acini that may not be possible due to the limiting imaging-depth of the confocal objectives. For both of these methods, the time and temperature at which fixation and permeabilization are performed are critical, especially given that TUM622 cells are embedded in a dense matrix (>90% basement membrane matrix) in contrast to many other 3D cultures where matrix density is much lower. Therefore, attention to standardized and consistent fixation and processing is necessary to obtain replicative results.

Using this system as a platform, one can then investigate how cell-intrinsic changes in the tumor cells, as well as cell-extrinsic changes in the tumor microenvironment, influence epithelial architecture and model early events involved in carcinoma formation. For example, the roles of oncogenes or tumor suppressor genes in regulating tumor tissue architecture could be studied by gain- or loss-of-function experiment targeting the gene of interest in the tumor cells. Indeed, we demonstrated that over-expression of SOX2, which is commonly observed in LUSC, alters the phenotype of TUM622 cells as evidenced by a loss of hyperplasia in 3D and progression towards dysplastic growth9. On the other hand, one could compare normal versus cancer-associated fibroblasts in coculture settings, determine how matrix components or its stiffness impact acini growth/morphology/invasion, and if blocking certain cytokines could interfere with cell-cell communication and in turn affect tissue architecture and tumor progression. Importantly, all these assays could be performed in the presence or absence of certain therapeutic agents and be used as a tool to determine the drug response of LUSC cells with a multidimensional readout11. It is also important to note that this system is limited in regards to the pathways it could be used to interrogate, as only some but not all major signaling pathways regulate the growth and morphology of TUM622 organoids in culture (i.e., inhibition of Wnt but not Notch signaling affects acinar mophogenesis of TUM622 cells)9.

In summary, we demonstrate that this organoid system provides a unique platform for generating new insights into the dynamic interplay between LUSC cells and the tumor microenvironment during tumor progression. We anticipate that our model system will be a valuable platform for drug discovery and development. In this respect, screening novel anti-cancer therapeutics in a native tumor tissue context should aid in the selection and development of more effective therapeutics targeting LUSC.

Subscription Required. Please recommend JoVE to your librarian.

Disclosures

The authors are employees and shareholders of Pfizer Inc.

Acknowledgments

We thank Magali Guffroy, John Kreeger, and Stephani Bisulco of the Pfizer-Oncology Histopathology and Biomarker group for pathology/histology support and Michael Arensman for critical review of the manuscript. We also thank the Pfizer Postdoctoral Program and the Oncology R&D group, specifically Robert Abraham, Puja Sapra, Karen Widbin and Jennifer Tejeda for their support of the program.

Materials

Name Company Catalog Number Comments
Bronchial Epithelial Growth Medium Lonza CC-3170 BEGM
Cell Strainer 40um ThermoFisher 352340 For passing TUM622 cells
Cleaved Caspase 3 antibody Cell Signaling Technology 9661 (RRID:AB_2341188) Rabbit
CoolRack CFT30 Biocision BCS-138 For 3D culture
CoolSink XT96F Biocision BCS-536 For 3D culture
Cultrex 3D Cell Harvesting Kit Bio-Techne 3448-020-K
Cultrex (preferred for co-culture) Bio-Techne 3443-005-01 For 3D culture
CXCR4 antibody Abcam Ab124824 (RRID:AB_10975635) Rabbit
E-cadherin antibody BD Biosciences 610182 (RRID:AB_397581) Mouse
GelCount Oxford Optronix For Acini counts and measurements
GM130 antibody BD Biosciences 610822 (RRID:AB_398141) Mouse
Goat Serum Vector Labs S1000 (RRID:AB_2336615) For Immunofluorescence
Heat-inactivated FBS Gibco 10082-147 For CAFs
Histology sample gel Richard Allan Scientific HG-4000-012 For Immunofluorescence
Integrin alpha 6 antibody Millipore Sigma Mab1378 (RRID:AB_2128317) Rat
Involucrin antibody Abcam Ab68 (RRID:AB_305656) Mouse
Ki67 antibody Abcam Ab15580 (RRID:AB_443209) Rabbit
Lab-Tec II chambered #1.5 German Coverglass System Nalge Nunc International 155379 (2) For 3D culture
Lab-Tec II chambered #1.5 German Coverglass System Nalge Nunc International 155409 (8) For 3D culture
L-Glutamine Gibco 25030-081 For CAFs
Matrigel (preferred for mono-culture) Corning 356231 For 3D culture
p63 antibody Cell Signaling Technology 13109 (SRRID:AB_2637091) Rabbit
Pen/Strep Gibco 15140-122 For CAFs
ReagentPack Subculture Reagents Lonza CC-5034 For TUM622 cell dissociation
RPMI ThermoFisher 11875-093 For CAFs
Sox2 antibody Cell Signaling Technology 3579 (RRID:AB_2195767) Rabbit
TrypLE Express Gibco 12604-021 For CAF dissociation
Vi-Cell Bechman Coulter Automatic cell counter
Vimentin antibody Abcam Ab92547 (RRID:AB_10562134) Rabbit
β-catenin antibody Cell Signaling Technology 2677s (RRID:AB_1030943) Mouse

DOWNLOAD MATERIALS LIST

References

  1. Cancer Genome Atlas Research Network. Comprehensive genomic characterization of squamous cell lung cancers. Nature. 489 (7417), 519-525 (2012).
  2. Nelson, C. M., Bissell, M. J. Of Extracellular Matrix, Scaffolds, and Signaling: Tissue Architecture Regulates Development, Homeostasis, and Cancer. Annual Review of Cell and Developmental Biology. 22 (1), 287-309 (2006).
  3. Quail, D. F., Joyce, J. A. Microenvironmental regulation of tumor progression and metastasis. Nature Medicine. 19 (11), 1423-1437 (2013).
  4. Balkwill, F. R., Capasso, M., Hagemann, T. The tumor microenvironment at a glance. Journal of Cell Science. 125 (23), 5591-5596 (2012).
  5. Schmeichel, K. L., Bissell, M. J. Modeling tissue-specific signaling and organ function in three dimensions. Journal of Cell Science. 116 (Pt 12), 2377-2388 (2003).
  6. Wu, X., Peters-Hall, J. R., Bose, S., Pena, M. T., Rose, M. C. Human bronchial epithelial cells differentiate to 3D glandular acini on basement membrane matrix. American Journal of Respiratory Cell and Molecular Biology. 44 (6), 914-921 (2011).
  7. Godugu, C., Singh, M. AlgiMatrix-Based 3D Cell Culture System as an In Vitro Tumor Model: An Important Tool in Cancer Research. Methods in Molecular Biology. 1379, 117-128 (2016).
  8. Amann, A., et al. Development of an innovative 3D cell culture system to study tumour--stroma interactions in non-small cell lung cancer cells. PLoS One. 9 (3), e92511 (2014).
  9. Chen, S., et al. Cancer-associated fibroblasts suppress SOX2-induced dysplasia in a lung squamous cancer coculture. Proceedings of the National Academy of Sciences of the United States of America. 115 (50), E11671-E11680 (2018).
  10. Damelin, M., et al. Delineation of a cellular hierarchy in lung cancer reveals an oncofetal antigen expressed on tumor-initiating cells. Cancer Research. 71 (12), 4236-4246 (2011).
  11. Sapra, P., et al. Long-term tumor regression induced by an antibody-drug conjugate that targets 5T4, an oncofetal antigen expressed on tumor-initiating cells. Molecular Cancer Therapeutics. 12 (1), 38-47 (2013).
  12. Fischer, A. H., Jacobson, K. A., Rose, J., Zeller, R. Paraffin embedding tissue samples for sectioning. Cold Spring Harbor Protocols. 2008, pdb.prot4989 (2008).

Tags

Tumor-stroma Interactions Tumor Morphology 3D Context Malignant Cells Tumor-stroma Cell Interactions Drug Treatment Basement Membrane Matrix Plastic Pipettes HEPES Buffer Trypsin/EDTA Trypsin Neutralization Buffer Centrifuge Tubes
Multidimensional Coculture System to Model Lung Squamous Carcinoma Progression
Play Video
PDF DOI DOWNLOAD MATERIALS LIST

Cite this Article

Chen, S., Giannakou, A., Golas, J.,More

Chen, S., Giannakou, A., Golas, J., Geles, K. G. Multidimensional Coculture System to Model Lung Squamous Carcinoma Progression. J. Vis. Exp. (157), e60644, doi:10.3791/60644 (2020).

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