Waiting
로그인 처리 중...

Trial ends in Request Full Access Tell Your Colleague About Jove

Developmental Biology

Three-dimensional Angiogenesis Assay System using Co-culture Spheroids Formed by Endothelial Colony Forming Cells and Mesenchymal Stem Cells

Published: September 18, 2019 doi: 10.3791/60032
* These authors contributed equally

Summary

Three-dimensional co-culture spheroid angiogenesis assay system is designed to mimic the physiologic angiogenesis. Co-culture spheroids are formed by two human vascular cell precursors, ECFCs and MSCs, and embedded in collagen gel. The new system is effective for evaluating angiogenic modulators, and provides more relevant information to the in vivo study.

Abstract

Studies in the field of angiogenesis have been aggressively growing in the last few decades with the recognition that angiogenesis is a hallmark of more than 50 different pathological conditions, such as rheumatoid arthritis, oculopathy, cardiovascular diseases, and tumor metastasis. During angiogenesis drug development, it is crucial to use in vitro assay systems with appropriate cell types and proper conditions to reflect the physiologic angiogenesis process. To overcome limitations of current in vitro angiogenesis assay systems using mainly endothelial cells, we developed a 3-dimensional (3D) co-culture spheroid sprouting assay system. Co-culture spheroids were produced by two human vascular cell precursors, endothelial colony forming cells (ECFCs) and mesenchymal stem cells (MSCs) with a ratio of 5 to 1. ECFCs+MSCs spheroids were embedded into type I collagen matrix to mimic the in vivo extracellular environment. A real-time cell recorder was utilized to continuously observe the progression of angiogenic sprouting from spheroids for 24 h. Live cell fluorescent labeling technique was also applied to tract the localization of each cell type during sprout formation. Angiogenic potential was quantified by counting the number of sprouts and measuring the cumulative length of sprouts generated from the individual spheroids. Five randomly-selected spheroids were analyzed per experimental group. Comparison experiments demonstrated that ECFCs+MSCs spheroids showed greater sprout number and cumulative sprout length compared with ECFCs-only spheroids. Bevacizumab, an FDA-approved angiogenesis inhibitor, was tested with the newly-developed co-culture spheroid assay system to verify its potential to screen anti-angiogenic drugs. The IC50 value for ECFCs+MSCs spheroids compared to the ECFCs-only spheroids was closer to the effective plasma concentration of bevacizumab obtained from the xenograft tumor mouse model. The present study suggests that the 3D ECFCs+MSCs spheroid angiogenesis assay system is relevant to physiologic angiogenesis, and can predict an effective plasma concentration in advance of animal experiments.

Introduction

Approximately 500 million people worldwide are expected to benefit from angiogenesis-modulating therapy for vascular malformation-associated diseases such as rheumatoid arthritis, oculopathy, cardiovascular diseases, and tumor metastasis1. Thus, the development of drugs that control angiogenesis has become an important research area in the pharmaceutical industry. During the drug development process, in vivo animal study is necessary to explore the effects of drug candidates on physiologic functions and systemic interactions between organs. However, ethical and cost issues have increased the concerns regarding animal experiments2. Therefore, improved in vitro assay systems are needed to obtain more accurate and predictable data leading to the better decision-making before animal experiments. Current in vitro angiogenesis assays usually measure proliferation, invasion, migration, or tubular structure formation of endothelial cells (ECs) seeded in two-dimensional (2D) culture plates3. These 2D angiogenesis assays are quick, simple, quantitative, and cost-effective, and have significantly contributed to the discovery of angiogenesis-modulating drugs. However, several issues remain to be improved.

Such 2D in vitro assay systems cannot reflect complex multi-step events of angiogenesis that occurs in in vivo physiologic conditions, leading to inaccurate results that cause discrepancies between in vitro assay data and clinical trial outcomes4. 2D culture conditions also induce the change of cellular phenotypes. For example, after proliferation in 2D culture plates, ECs have a weak cellular phenotype as manifested by reduced expression of CD34 and several signals that govern cellular responses5,6. To overcome the limitations of 2D culture-based angiogenesis assay systems, three-dimensional (3D) spheroid angiogenesis assay systems have been developed. Sprouting followed by tubular structure formation from spheroids formed by ECs reflect in vivo neo-vascularization processes7,8. Thus, the 3D spheroid angiogenesis assay has been considered an effective assay system for screening potential pro- or anti-angiogenesis drugs.

Most 3D spheroid angiogenesis assays utilize only ECs, mainly human umbilical vein endothelial cells (HUVECs) or human dermal microvascular endothelial cells (HDMECs) to focus on the cellular response of ECs during angiogenesis. However, blood capillaries are composed of two cell types: ECs and pericytes. Elaborating bi-directional interaction between ECs and pericytes is critical for proper vascular integrity and function. Several diseases, such as hereditary stroke, diabetic retinopathy, and venous malformation, are associated with altered pericyte density or decreased pericyte attachment to the endothelium9. Pericytes are also known as a key element of the angiogenic process. Pericytes are recruited to stabilize newly-formed vessel structures by ECs. In this regard, mono-culture spheroid angiogenesis assay does not incorporate pericytes7,10. Therefore, co-culture spheroids formed by ECs and pericytes may provide a valuable approach to more closely mimic physiologic angiogenic events.

The present study aimed to develop a 3D co-culture spheroid angiogenesis assay with a combination of human endothelial colony forming cells (ECFCs) and mesenchymal stem cells (MSCs) to more closely reflect in vivo angiogenesis. Co-culture spheroid system as an in vitro representation assembly of a normal blood vessel was first established by Korff et al. in 200111. They combined HUVECs and human umbilical artery smooth muscle cells (HUSMCs), and demonstrated that co-culture of two mature vascular cells decreased the sprouting potential. Mature ECs (HUVECs) are known to progressively lose their ability to proliferate and differentiate, which negatively affects their angiogenesis responses12,13. Mature perivascular cells (HUSMCs) can cause endothelial cell inactivation through the abrogation of the vascular endothelial growth factor (VEGF) responsiveness11. The main difference between Korff’s and our co-culture spheroid system is the cell types used. We applied two vascular precursors, ECFCs and MSCs, to establish a proper angiogenesis assay system to screen and investigate pro-or-anti-angiogenic agents. ECFCs are the precursor of ECs. ECFCs have robust proliferation capacity compared with mature ECs14, which enable to overcome the limitation of ECs. ECFCs contribute to new vessel formation in many post-natal pathophysiologic conditions15,16,17. MSCs are pluripotent stem cells that have the capacity to differentiate into pericytes, thereby contributing to angiogenesis18,19.

In previous reports, ECFCs and MSCs showed synergistic effects on in vitro tube formation20, in vivo neo-vascularization21,22, and improved reperfusion of ischemic tissues23,24. In the present study, ECFCs and MSCs were used to form co-culture spheroids and embedded in type I collagen gel to reflect an in vivo 3D environment. Collagen is considered as a major constituents of the extracellular matrix (ECM) surrounding ECs25. The ECM plays a critical role in regulating cell behavior26. The assay protocol proposed here can be easily and quickly carried out within two days using common laboratory techniques. For effective cell tracking during the sprouting process, each cell type can be fluorescently labeled and monitored using a real-time cell recorder. The newly-established 3D co-culture spheroid angiogenesis assay system is designed to increase sensitivity for evaluating potential angiogenic modulators and to provide predictable information in advance of in vivo study.

Subscription Required. Please recommend JoVE to your librarian.

Protocol

Human ECFCs were isolated from human peripheral blood as described in a previous report27. Briefly, the mononuclear cell layer was separated from the whole blood using the Ficoll-Paque Plus, and cultured in the proper medium until the endothelial-like colonies were appeared. Colonies were collected and ECFCs were isolated using CD31-coated magnetic beads. MSCs were isolated from the adherent mononuclear cell (MNC) fraction of human adult bone marrow. The study protocol was approved by the institutional review board of Duksung Women’s University (IRB No. 2017-002-01).

1. Cell Culture

  1. Preparing ECFCs and MSCs medium and coat plates
    1. Prepare endothelial cell growth medium MV2 (ECGM-MV2, except hydrocortisone) supplemented with 10% fetal bovine serum (FBS) and 1% glutamine-penicillin-streptomycin (GPS).
    2. Prepare mesenchymal stem cell growth medium-2 (MSCGM-2) containing 10% FBS and 1% GPS.
    3. For ECFC culture, coat the cell culture plates with 1% gelatin solution. To prepare 1% gelatin solution, dissolve 1 g of gelation powder in 500 mL of PBS with the magnetic stirrer, and sterilize with autoclave. Plates can be coated with 3 mL/60 mm, 5 mL/100 mm, or 15 mL/150 mm of 1% gelatin solution. Incubate coated plates for 15 min in a cell culture incubator (37 °C and 5% CO2). After then, remove the remaining 1% gelatin solution by aspiration and wash the coated plates one time with PBS.
  2. Growing ECFCs and MSCs
    1. Seed 1 x 106 ECFCs on 1% gelatin-coated 150 mm plates, and grow using ECGM-MV2 (10% FBS, 1% GPS) in a cell culture incubator (37°C and 5% CO2) to 80-90% confluency. Use ECFCs at passage numbers 7-10 to obtain consistent results.
    2. Seed 1 x 106 MSCs on non-coated 150 mm plates, and grow using MSCGM-2 in a cell culture incubator (37°C and 5% CO2) to 80-90% confluency. Use MSCs at passage numbers 7-10 to obtain consistent results.

2. Preparation of 1.2% w/v Methylcellulose Solution

  1. Measure 6 g of methyl cellulose in a 500 mL glass bottle and sterilize in an autoclave.
  2. Heat ECGM-MV2 (without FBS and GPS) at 60 °C for 20 min, and add 250 mL of heated ECGM-MV2 to sterilized methyl cellulose powder. To maintain sterile conditions, carry out the process inside the flow hood.
  3. Add a sterilized magnetic stir bar and mix the solution for 20 min on the magnetic stirrer until the methyl cellulose is thoroughly wetted and evenly dispersed with no lumps. Add 250 mL of cold (4 °C) ECGM-MV2 (without FBS and GPS) under the sterile condition and mix for an additional 10 min on the magnetic stirrer. Then, chill the solution in a refrigerator (4 °C) overnight.
    NOTE: Methylcellulose is a carbohydrate polymer that dissolve well in cool temperatures due to swelling and subsequent hydration.
  4. The next day, aliquot the solution in a 50 mL tube and centrifuge at 5,000 x g for 3 h at 4 °C. Take the clear viscous supernatant solution and store at 4 °C until use for up to 3-6 months.
    NOTE: The sediment may contain residual cellulose fiber, so gently take the supernatant solution leaving behind at least 5 mL of volume. This volume can be adjusted according to experimental conditions.

3. Generation and Embedding of ECFCs-only, MSCs-only, and ECFCs-MSCs Spheroids

Day 1

  1. Preparing ECFCs and MSCs cell suspension
    1. Wash the 80-90% confluent ECFCs and MSCs by adding phosphate buffered saline (PBS) without calcium and magnesium, and aspirate.
    2. To detach ECFCs and MSCs from the culture plate, incubate them with 0.05% trypsin-EDTA for 3 min and 5 min in a cell culture incubator (37°C and 5% CO2), respectively, in a cell culture incubator.
    3. Inactivate trypsin by adding DMEM medium containing 10% FBS and 1% GPS. Pipette the cells up and down to create a single cell suspension.
    4. Sediment the cells by centrifuging at 282 x g for 5 min at RT.
    5. Remove the supernatant, and re-suspend the cells in the respective medium.
  2. Labeling ECFCs and MSCs with fluorescent dye
    NOTE: Perform cell membrane labelling of ECFCs with PKH67 (green) and MSCs with PKH26 (red) fluorescent dye following the manufacturer’s instructions with slightly modifications as follows.
    1. Wash the cells two times with serum free medium to remove FBS.
      NOTE: Proteins and lipids in FBS reduces the effective dye concentration for labelling cells by binding.
    2. Count ECFCs and MSCs using a hemocytometer under the microscope, and transfer 3 x 106 ECFCs and 2 x 106 MSCs into 2 mL micro-tubes.
      NOTE: These are the optimum cell densities for labelling cells with dyes. Use of a large number of cells causes poor and heterogeneous staining, whereas use of too few cells yields poor cells recovery.
    3. Centrifuge the tubes at 100 x g for 5 min at RT to obtain a cell pellet. Carefully aspirate the supernatant leaving no more than 15-25 µL of residual volume.
    4. Re-suspend ECFCs and MSCs in 250 µL of Diluent C from the dye kit. Gently pipette the cell suspension to ensure complete dispersion.
      NOTE: Diluent C is a physiologic salt, and may reduce the staining efficiency of dye by forming micelles. Therefore, do not let cells stand in diluent C for long, and do not vortex the tubes.
    5. Prepare the staining solution for ECFCs by adding 5 µL of PKH67 to 250 µL of Diluent C (final concentration of 20 µM) and for MSCs by adding 3 µL of PKH26 to 250 µL of Diluent C (final concentration of 12 µM ).
      NOTE: The final dye concentrations are different from the manufacturer’ recommendations. High concentration would lead to cell clumping and cell toxicity. Low concentration would not be sufficient for staining.
    6. Add 250 µL of ECFCs cell suspension to 250 µL of PKH67 dye solution, and 250 µL of MSCs to 250 µL of PKH26 dye solution. Immediately mix the cells with dyes by pipetting up and down and incubate for 5 min at room temperature. For better results, cover the tube with aluminum foil and place on a shaker to ensure sufficient mixing during staining.
    7. Stop the staining process by adding 0.5 mL of FBS to the stained cell suspension, and then incubate for 1 min to allow FBS to bind excess unbound dye. Sediment the stained cells by centrifuging at 100 x g for 5 min.
      NOTE: ECFCs and MSCs pellets are light yellow and pink, respectively, after staining.
    8. Carefully remove the supernatant and wash the cells two times with complete medium to remove excess dye. Centrifuge the tubes at 100 x g for 5 min and re-suspend the pellet in 2 mL of each complete medium.
  3. Generating ECFC, MSC, or ECFC+MSC spheroids
    NOTE: Apply the hanging drop technique to generate spheroids as described previously28. The steps for the preparation of spheroids are given below.
    1. Count the stained ECFCs and MSCs.
    2. Suspend stained ECFCs, MSCs or ECFCs+MSCs in ECGM-MV2 containing 20% Methylcellulose solution and 5% FBS. Prepare at a cell density of 4 x 104 cells/mL for ECFCs or MSCs (25 µL of cell suspension contains 1,000 cells). For two cell suspension of ECFCs and MSCs, prepare at a cell density of 2 x 104 cells/mL ECFCs and 0.4 x 104 cell/mL MSCs (25 µL contains 500 ECFCs and 100 MSCs).
      NOTE: Ratio of ECFCs to MSCs should not exceed 5:1. A larger number of MSCs could lead to excessive migration and irregular sprout morphology. A smaller number of MSCs could cause poor interaction between cells, yielding poor sprouting.
    3. Prepare a hydration unit by adding 15 mL of PBS into the bottom of a 150 mm culture plate.
    4. Transfer the cell suspension into the sterilized polystyrene rectangular reservoir for the use of multichannel pipette. Disperse the cells suspension evenly by pipetting.
    5. Deposit 25 µL drops of cell suspension onto the cover of a 150 mm culture plate using a multi-channel pipette (approximately 100 drops/cover of the 150 mm plate). Invert the cover over a PBS-filled hydration unit and incubate in a cell culture incubator for 24 h.
      NOTE: Methylcellulose solution provides a proper viscosity to the suspension solution. When the hanging drop method was performed without methylcellulose solution, drops were easily slipped down when the cover was inverted (Supplemental Figure 1). Furthermore, after overnight incubation, the spheroids were not perfectly formed without methylcellulose solution. This result indicate that proper viscosity by methylcellulose solution is necessary to form round shape of the spheroids.

Day 2

  1. Embeding spheroids in neutralized collagen gel
    1. Warm FBS and ECGM-MV2 (without FBS and GPS) in a water bath (37 °C). Place cold methylcellulose solution on a work bench to bring to room temperature.
    2. Place a 24-well plate in a cell culture incubator (37 °C) for warming.
    3. Cut the pointed 1 mL pipette tips 3-5 mm to aspirate spheroids and viscous suspensions, such as methylcellulose solution and type I collagen, comfortably and accurately.
    4. Prepare 3 mg/mL neutralized type I collagen gel on ice following the type I collagen gel manufacturer’s instruction.
      NOTE: Neutralization should be performed on ice to avoid gelation of collagen at room temperature. For one well of a 24-well plate, 500 µL of neutralized collagen is needed. Always prepare 1 mL extra volume of collagen. Collagen gel can be used up to 2-3 h after neutralization if kept on ice.
    5. Harvest the spheroids by rinsing the cover containing spheroids with 5 mL of PBS. Collect spheroid-suspended solution in a 50 mL conical tube. Re-rinse the cover with 5 mL of PBS to obtain the remaining spheroids.
      NOTE: During harvest, closely observe to confirm the existence of spheroids. For more visual inspection, transfer about 50-100 µL of spheroid-suspended solution on the slide glass, and check the round shape of the spheroids under the microscope.
    6. Sediment the spheroids by centrifuging at 282 x g for 5 min. Discard the supernatant carefully without disturbing the spheroids leaving behind not more than 100-200 µL of residual volume.
    7. Gently tap on the wall of the tube so that spheroids are freely suspended in the remaining 100-200 µL residual solution.
    8. Add ECGM-MV2 containing 5% FBS and 40% methylcellulose solution to the spheroid-containing tube. The volume added is determined based on approximately 100 spheroids/mL. Gently mix the spheroid suspension by pipetting with a blunt 1 mL pipette tip.
      NOTE: Methylcellulose solution is widely used as a suspending agent that does not allow spheroids to sediment. FBS concentration should be 5%. Higher FBS concentration causes abnormal sprouting due to excessive growth factors. Lower FBS concentration cannot maintain healthy conditions for cells.
    9. Mix spheroid-suspension solution and neutralized-type I collagen gel (3 mg/mL) at a 1:1 ratio on ice. Use a blunt 1 mL pipette tip to avoid breaking of spheroids.
      NOTE: 1 mL of the spheroids-suspending collagen gel solution is required for one well of a 24-well plate. Make an extra mixed suspension solution if possible.
    10. If the angiogenic properties of any agent(s) or chemical(s) need to be tested, transfer 1 mL of the spheroid-suspending collagen gel solution in a 1 mL micro-tube and add agent(s) or chemical(s) followed by gentle mixing with a blunt 1mL pipette tip.
      NOTE: The volume of agent and chemical can sum to 200 μL, which dilutes the type I collagen gel concentration from 1.5 mg/mL to 1.25 mg/mL, but does not affect type I collagen gel polymerization. Add same volume of vehicle to the control spheroids.
    11. Add the spheroid-suspending collagen gel solution into wells of a pre-warmed 24-well plate (0.9 mL/well) by pipetting, and then incubate for 30 min in a cell culture incubator for polymerization.
      NOTE: During the spheroid embedding process, do not disturb the gel by agitating the plate.
    12. Cover the spheroid-suspending collagen gel by adding 100 µL of ECGM-MV2 containing 2.5% FBS with/without VEGF. For ECFC-only spheroids, stimulate the cells with exogenous addition of VEGF (final concentration of 50 ng/mL) to form proper sprouts. ECFC+MSC spheroids do not require exogenous stimulation by any growth factors.
    13. Place the plate in a real-time cell recorder installed in a cell culture incubator (37 °C and 5% CO2), and randomly focus on 5-10 spheroids (10x objective lens). Monitor the sprouting formation of each fluorescence-labeled spheroids every 1 h for 24 h without any disturbance.

4. Quantitate Spheroid Sprouting

  1. Import the image files to ImageJ software to quantitate the number of sprouts and measure the length of each sprout. For co-culture spheroids, label ECFCs with green fluorescent dye before making spheroids. Then, measure the number and length of sprouts stained with green fluorescence (Supplemental Figure 2). Five randomly-selected spheroids were quantified per experimental group.
    NOTE: Sprouts are collaboratively elongated structures formed by several ECFCs extending out of spheroids. Sprout length is measured as the length from the point of sprouts origin from the surface of the spheroids to the tip of sprouts.
  2. Express the values as means ± SEM of at least three independent experiments. Determine the statistically significant difference by one-way ANOVA for multiple comparisons or Student’s t-test for paired comparisons.
    NOTE: P ≤ 0.05 is considered statistically significant.

Subscription Required. Please recommend JoVE to your librarian.

Representative Results

Comparison experiments were performed using mono-culture spheroids (ECFCs-only) and co-culture spheroids (ECFCs+MSCs) to examine whether MSCs play a considerable role in ECFCs-mediated angiogenesis. Sprouting formation from each spheroid was monitored for 24 h by a real-time cell recorder that could capture the progression of angiogenic sprouting from spheroids. Angiogenic potential was quantified by counting the number of sprouts and measuring the cumulative length of sprouts generated from individual spheroids. Five randomly-selected spheroids per experimental group were analyzed. For ECFCs+MSCs spheroids, number of sprouts and cumulative sprout length were significantly higher compared with those of ECFCs-only spheroids at all time-points (Figure 1A-C). Sprout number and length of ECFCs+MSCs spheroids increased continuously for 12 h, but number and length of ECFCs-only spheroids increased for 6 h and did not change at later time-points (Figure 1B,C). In addition, sprouts formed by ECFCs+MSCs spheroids were thicker and more durable than those formed by ECFCs-only spheroids with/without VEGF treatment (Figure 1A, and Supplemental Video 1A,B,D). MSCs-only spheroids did not form sprouts but showed individual migration of MSCs outside of spheroids (Figure 1A and Supplemental Video 1C). These results demonstrate the significant contribution of MSCs, pericyte precursors, to the cellular angiogenesis of ECFCs. MSCs are known to secrete various growth factors29 that may stimulate ECFCs to form sprouts and tubular structures.

ECFCs were labelled with green-fluorescent dye and MSCs were labeled with red-fluorescent dye before combining to generate ECFCs+MSCs spheroids. Together with real-time recording , this live cell fluorescence labeling technique can track the cellular movements of ECFCs and MSCs during sprout formation. Fluorescent imaging showed that ECFCs-mediated sprout structures were covered with MSCs (Figure 2 and Supplemental Video 2). This suggest that combined MSCs function as perivascular cells during sprout formation, which enhance sprout stability and durability by the tight association between two vascular cells.

The newly-developed co-culture spheroid assay system was tested with bevacizumab, an FDA-approved angiogenesis inhibitor, to verify its potential to screen anti-angiogenic drugs. ECFCs+MSCs spheroids pretreated with bevacizumab showed decreased sprout number and cumulative sprout length in a dose-dependent manner compared with control ECFCs+MSCs spheroids (Figure 3A,B). In parallel experiments, ECFCs-only spheroids pretreated with bevacizumab followed by stimulation with VEGF (50 ng/mL) also showed decreased VEGF-induced sprout number and cumulative sprout length in a dose-dependent manner (Figure 3C,D). Of note, the IC50 values of bevacizumab for inhibiting cumulative sprout length in ECFCs+MSCs spheroids was 46 times greater than that in ECFCs-only spheroids (Table 1). This result strongly implies that higher concentration is needed to inhibit physiologically-relevant vascular formation by ECFCs and MSCs compared with the concentration needed to inhibit only EC-mediated vascular formation.

Next, a xenograft tumor mouse model was performed with bevacizumab treatment to reveal which spheroid system provide predictable data correlating with in vivo effective plasma concentration. Human-derived glioblastoma U87MG-Red-FLuc cell line was subcutaneously injected to immune-deficient mice. After confirming tumor formation at 1 week, mice were randomly divided into 3 groups that received different treatments: control (0 mg/kg), low dose (1 mg/kg), and high dose (30 mg/kg). Tumor growth was significantly inhibited in the 30 mg/kg-treated group compared with control group (Supplemental Figure 3A). The 1 mg/kg-treated group showed a tendency for tumor decrease, but there was no statistical difference compared with control group. Mouse plasma concentration at 3 week of bevacizumab treatment was 568.0±40.62 µg/mL at 30 mg/kg and 38.1±0.72 µg/mL at 1 mg/kg (Supplemental Figure 3B). Notably, plasma concentration of bevacizumab showing effective inhibition (568.0±40.62 µg/mL at 30 mg/kg for 3 week treatment) was closely achieved by ECFCs+MSCs spheroids (1261.5±214.49 µg/mL) but not ECFCs-only spheroids (27.0±9.97 µg/mL). Thus, the ECFCs+MSCs spheroid angiogenesis assay system can be considered as a suitable assay system for predicting effective plasma concentration in advance of animal experiments.

Figure 1
Figure 1: Sprout formation from ECFCs-only, MSCs-only, and ECFCs+MSCs spheroids. (A) Representative images of sprout formation from ECFCs-only, MSCs-only and ECFCs+MSC spheroids embedded in type I collagen gel at 0, 6, 12, 18, and 24 h (Scale bar = 100 µm). (B) Quantitative graph of sprout number formed from ECFCs-only and ECFCs+MSCs spheroids (mean±SEM, n = 3). (C) Quantitative graph of cumulative sprout length formed from ECFCs-only and ECFCs+MSCs spheroids (mean±SEM, n = 3). * indicates significant difference between ECFCs-only and ECFCs+MSCs spheroids at the same time-points (p ≤ 0.05). # indicates significant difference between groups indicated by a bracket (p ≤ 0.05). This figure has been modified from a previous pubication30. Please click here to view a larger version of this figure.

Figure 2
Figure 2: Localization of ECFCs and MSCs in sprout structures. Representative images showing locations of two cell types in sprout structures after 24 h. ECFCs and MSCs were fluorescently labelled with PKH67 (green) and PKH26 (red), respectively, and performed 3D ECFCs+MSCs spheroid angiogenesis assay. Arrows shows that MSCs were covered with ECFCs-mediated sprout structures (Scale bar = 100 µm). Please click here to view a larger version of this figure.

Figure 3
Figure 3: Inhibitory effect of bevacizumab on sprout formation from ECFCs+MSCs and ECFCs-only spheroids. ECFCs+MSCs and ECFCs-only spheroids were treated with bevacizumab, and sprout formation was monitored for 24 h. (B) Quantitative graph of cumulative sprout length from ECFCs+MSCs spheroids treated with bevacizumab (mean±SEM, n = 3). (C) Quantitative graph of sprout number from VEGF-stimulated ECFCs-only spheroids treated with bevacizumab (mean±SEM, n = 3). (D) Quantitative graph of cumulative sprout length from VEGF-stimulated ECFCs-only spheroids treated with bevacizumab (mean±SEM, n = 3). * indicates significant difference from control group (white bar) (p ≤ 0.05). # indicates significant difference from VEGF-treated group (black bar) (p ≤ 0.05). Please click here to view a larger version of this figure.

Time
(h)
IC50 (µg/mL) of Avastin p-value
ECFCs-only spheroid ECFCs+MSCs spheroid
6 94.62 ± 38.53 3058.21 ± 373.31 0.003
12 58.61 ± 17.80 2006 ± 484.73 0.015
18 83.38 ± 54.54 1509.51 ± 483.88 0.042
24 27.04 ± 9.97 1261.51 ± 214.49 0.0045

Table 1: IC50 values of bevacizumab for inhibition of cumulative sprout length in either ECFCs-only or ECFCs+MSCs spheroids. ECFCs-only spheroids were treated with bevacizumab followed by stimulation with VEGF (50 ng/mL), which is required for sprout formation from ECFCs-only spheroids. ECFCs+MSCs spheroids were treated with bevacizumab without VEGF stimulation. Both spheroids embedded into type I collagen gel, and sprout formation from each spheroid was observed for 24 h using a real-time cell recorder. Data are represented as mean ± SEM (n = 3).

Supplemental Figure 1
Supplemental Figure 1: Please click here to view a larger version of this figure.

Supplemental Figure 2
Supplemental Figure 2: Please click here to view a larger version of this figure.

Supplemental Figure 3
Supplemental Figure 3: Please click here to view a larger version of this figure.

Supplemental Video 1A
Supplemental Video 1A: Please click here to view this video. (Right-click to download.)

Supplemental Video 1B
Supplemental Video 1B: Please click here to view this video. (Right-click to download.)

Supplemental Video 1C
Supplemental Video 1C: Please click here to view this video. (Right-click to download.)

Supplemental Video 1D
Supplemental Video 1D: Please click here to view this video. (Right-click to download.)

Supplemental Video 2
Supplemental Video 2: Please click here to view this video. (Right-click to download.)

Subscription Required. Please recommend JoVE to your librarian.

Discussion

The present study introduce an improved in vitro angiogenesis assay system utilizing co-culture spheroids formed by two human vascular cell progenitors, ECFCs and MSCs. Co-culture spheroid system can mimic in vivo vascular sprout formation, which is accomplished by interaction and incorporation between endothelial cells and pericytes. Compared to other in vitro angiogenesis assays that reflect only ECs-mediated angiogenesis, this co-culture assay system is more representative of the multistep cascade of physiologic angiogenesis including cellular interaction, sprouting, tube formation, and vessel maturation. This newly-established assay system also resembles the in vivo extracellular microenvironment by seeding spheroids into type I collagen gel. We suggest that the 3D co-culture spheroid angiogenesis assay system is reliable, repeatable, easily quantifiable, and most importantly physiologically relevant.

While performing the co-culture spheroid assay, it is essential to embed spheroids into gel with appropriate concentration of neutralized type I collagen and FBS. The best final concentration of neutralized type I collagen is 1.5 mg/mL, and the percentage of FBS is 2.5%. In the preliminary experiments, higher concentrations of collagen resulted in stiff gel and hampered the quality and quantity of sprouts originating from spheroids. Lesser concentrations of collagen yielded soft and fragile gel that could not maintain the integrity of spheroids. Similarly, higher amounts of FBS caused plethoric sprouting from spheroids at the basal level irrespective of angiogenic factor stimulation. Lower FBS concentration led to poor conditions of cells. For consistent and reproducible results, a proper number of spheroid should be embedded (about 50 spheroids/well). More than 50 spheroids/well could lead to close proximity of spheroids within the well, which may abnormally affect the quality and quantity of sprouts generated.

It is critical to maintain type I collagen in chilled conditions during neutralization and mixing steps with the spheroid suspension because collagen can clot at room temperature, resulting in an irregular matrix. While mixing the spheroid suspension with collagen solution, it is recommended to use 1 mL pipette tip with 3-5 mm cut at the end to make wide hole. This enables easy handling of the viscous collagen solution and protects spheroids from rupture. Agitation of the plate after embedding spheroid-suspended collagen solution could disturb the integrity of the gel and result in breakage that may hamper normal sprout generation.

In co-culture spheroid assay using ECFCs and MSCs, 5:1 ratio should be maintained. Use of a larger number of MSCs causes spreading around the spheroid due to the migratory phenotype of MSCs, which can affect ideal sprout generation. A smaller number of MSCs is insufficient to stimulate ECFCs sprouting and cover the sprouts properly. In addition, it is essential to check cell conditions during growth. If poor sprouting is observed, it is recommended to use another healthy passages of the cells. For better outcome, use of ECFCs and MSCs at passage less than 10 is strongly recommended.

Here, we present an improved 3D angiogenesis assay system using co-culture spheroids that closely capture in vivo angiogenesis. Compared with 2D single cell assay systems, this co-culture spheroid assay system reflect more faithfully cellular responses between two vascular cell types to form tubular structures in physiologic conditions. However, this system remains oversimplified compared to the complex multi-step process of in vivo angiogenesis. Vascular generation in vivo usually occurs through multiple interaction of various other cell types, including epithelial cells, fibroblasts, immune cells, and also abundant ECM proteins. Future applications of this new system include introduction of other cell types and the ECM to more precisely reflect the physiological and/or pathological angiogenesis.

Subscription Required. Please recommend JoVE to your librarian.

Disclosures

The authors have nothing to disclose.

Acknowledgments

This research was supported by a grant (17172MFDS215) from Ministry of Food and Drug Safety, the National Research Foundation of Korea(NRF) grant funded by the Korea government(MSIP) (2017R1A2B4005463), and the Basic Science Research Program through the National Research Foundation of Korea(NRF) funded by the Ministry of Education (2016R1A6A1A03007648).

Materials

Name Company Catalog Number Comments
0.05% Trypsin EDTA (1x) Gibco 25300-054
Bevacizumab Roche NA Commercial name: Avastin
Dulbecco Modified Eagle Medium Gibco 11885-084 DMEM
Dulbeco's Phosphate buffered saline (10x) Gibco 21600-010 PBS (10x)
Dulbeco's Phosphate buffered saline (1x) Corning 21-031-CVR PBS (1x)
Endothelial cell Growth medium MV2 kit Promocell C-22121 ECGM-MV2
Fetal bovine serum (FBS) Atlas FP-0500A FBS
Gelatin BD Sciences 214340
L-Glutamine–Penicillin–Streptomycin Gibco 10378-016 GPS
Mesenchymal stem cell growth medium-2 Promocell C-28009 MSCGM-2
Methyl cellulose Sigma-Aldrich M0512
PKH26 Fluorescent Cell Linker Kits Sigma-Aldrich MINI26 PKH26
PKH67 Fluorescent Cell Linker Kits Sigma-Aldrich MINI67 PKH67
Sodium Hydroxide Sigma-Aldrich S5881
Type I collagen gel Corning 354236
Vascular endothelial growth factor A R&D 293-VE-010 VEGF

DOWNLOAD MATERIALS LIST

References

  1. Carmeliet, P. Angiogenesis in life, disease and medicine. Nature. 438 (7070), 932-936 (2005).
  2. Goodwin, A. M. In vitro assays of angiogenesis for assessment of angiogenic and anti-angiogenic agents. Microvascular research. 74 (2-3), 172-183 (2007).
  3. Staton, C. A., Reed, M. W., Brown, N. J. A critical analysis of current in vitro and in vivo angiogenesis assays. International journal of experimental pathology. 90 (3), 195-221 (2009).
  4. Lutolf, M. P., Gilbert, P. M., Blau, H. M. Designing materials to direct stem-cell fate. Nature. 462 (7272), 433-441 (2009).
  5. Fina, L., et al. Expression of the CD34 gene in vascular endothelial cells. Blood. 75 (12), 2417-2426 (1990).
  6. Delia, D., et al. CD34 expression is regulated reciprocally with adhesion molecules in vascular endothelial cells in vitro. Blood. 81 (4), 1001-1008 (1993).
  7. Korff, T., Augustin, H. G. Tensional forces in fibrillar extracellular matrices control directional capillary sprouting. Journal of Cell Science. 112 (19), 3249-3258 (1999).
  8. Korff, T., Augustin, H. G. Integration of endothelial cells in multicellular spheroids prevents apoptosis and induces differentiation. The Journal of cell biology. 143 (5), 1341-1352 (1998).
  9. Gaengel, K., Genové, G., Armulik, A., Betsholtz, C. Endothelial-mural cell signaling in vascular development and angiogenesis. Arteriosclerosis, thrombosis, and vascular biology. 29 (5), 630-638 (2009).
  10. Hutmacher, D. W. Biomaterials offer cancer research the third dimension. Nature Materials. 9 (2), 90-93 (2010).
  11. Korff, T., Kimmina, S., Martiny-Baron, G., Augustin, H. G. Blood vessel maturation in a 3-dimensional spheroidal coculture model: direct contact with smooth muscle cells regulates endothelial cell quiescence and abrogates VEGF responsiveness. FASEB Journal. 15 (2), 447-457 (2001).
  12. Gumkowski, F., Kaminska, G., Kaminski, M., Morrissey, L. W., Auerbach, R. Heterogeneity of mouse vascular endothelium. In vitro studies of lymphatic, large blood vessel and microvascular endothelial cells. Journal of Vascular Research. 24 (1-2), 11-23 (1987).
  13. Asif, A. R., Oellerich, M., Armstrong, V. W., Hecker, M., Cattaruzza, M. T-786C polymorphism of the NOS-3 gene and the endothelial cell response to fluid shear stress-a proteome analysis. Journal of Proteome Research. 8 (6), 3161-3168 (2009).
  14. Melero-Martin, J. M., et al. In vivo vasculogenic potential of human blood-derived endothelial progenitor cells. Blood. 109 (11), 4761-4768 (2007).
  15. Ingram, D. A., et al. Identification of a novel hierarchy of endothelial progenitor cells using human peripheral and umbilical cord blood. Blood. 104 (9), 2752-2760 (2004).
  16. Murasawa, S., Asahara, T. Endothelial progenitor cells for vasculogenesis. Physiology (Bethesda). 20, 36-42 (2005).
  17. Rafii, S., Lyden, D., Benezra, R., Hattori, K., Heissig, B. Vascular and haematopoietic stem cells: novel targets for anti-angiogenesis therapy. Nature Review Cancer. 2 (11), 826-835 (2002).
  18. Shi, S., Gronthos, S. Perivascular niche of postnatal mesenchymal stem cells in human bone marrow and dental pulp. Journal of Bone and Mineral Research. 18 (4), 696-704 (2003).
  19. Crisan, M., et al. A perivascular origin for mesenchymal stem cells in multiple human organs. Cell Stem Cell. 3 (3), 301-313 (2008).
  20. Lee, H., Kang, K. T. Advanced tube formation assay using human endothelial colony forming cells for in vitro evaluation of angiogenesis. Korean Journal of Physiology & Pharmacology. 22 (6), 705-712 (2018).
  21. Melero-Martin, J. M., et al. Engineering robust and functional vascular networks in vivo with human adult and cord blood-derived progenitor cells. Circulation Research. 103 (2), 194-202 (2008).
  22. Kang, K. T., Allen, P., Bischoff, J. Bioengineered human vascular networks transplanted into secondary mice reconnect with the host vasculature and re-establish perfusion. Blood. 118 (25), 6718-6721 (2011).
  23. Kang, K. T., Coggins, M., Xiao, C., Rosenzweig, A., Bischoff, J. Human vasculogenic cells form functional blood vessels and mitigate adverse remodeling after ischemia reperfusion injury in rats. Angiogenesis. 16 (4), 773-784 (2013).
  24. Kang, K. T., Lin, R. Z., Kuppermann, D., Melero-Martin, J. M., Bischoff, J. Endothelial colony forming cells and mesenchymal progenitor cells form blood vessels and increase blood flow in ischemic muscle. Scientific Reports. 7 (1), 770 (2017).
  25. Paupert, J., Sounni, N. E., Noel, A. Lymphangiogenesis in post-natal tissue remodeling: lymphatic endothelial cell connection with its environment. Molecular Aspects of Medicine. 32 (2), 146-158 (2011).
  26. Lu, P., Weaver, V. M., Werb, Z. The extracellular matrix: a dynamic niche in cancer progression. Journal of Cell Biology. 196 (4), 395-406 (2012).
  27. Melero-Martin, J. M., Bischoff, J. Chapter 13. An in vivo experimental model for postnatal vasculogenesis. Methods in Enzymology. 445, 303-329 (2008).
  28. Laib, A. M., et al. Spheroid-based human endothelial cell microvessel formation in vivo. Nature Protocols. 4 (8), 1202-1215 (2009).
  29. Leuning, D. G., et al. The cytokine secretion profile of mesenchymal stromal cells is determined by surface structure of the microenvironment. Scientific Reports. 8 (1), 7716 (2018).
  30. Shah, S., Kang, K. T. Two-Cell Spheroid Angiogenesis Assay System Using Both Endothelial Colony Forming Cells and Mesenchymal Stem Cells. Biomolecules & Therapeutics (Seoul). 26 (5), 474-480 (2018).

Tags

3D Angiogenesis Assay System Co-culture Spheroids Endothelial Colony Forming Cells Mesenchymal Stem Cells Physiologic Angiogenesis Angiogenic Modulators In Vivo Studies Vascular Progenitor Cells Cellular Interactions Sprouting Tubular Maturation Personalized Treatments Abnormal Angiogenesis-related Diseases Cancer Drug Development Trypsin-EDTA Cell Culture Incubator Complete DMEM Medium Single Cell Suspension Centrifugation
Three-dimensional Angiogenesis Assay System using Co-culture Spheroids Formed by Endothelial Colony Forming Cells and Mesenchymal Stem Cells
Play Video
PDF DOI DOWNLOAD MATERIALS LIST

Cite this Article

Shah, S., Lee, H., Park, Y. H.,More

Shah, S., Lee, H., Park, Y. H., Jeon, E., Chung, H. K., Lee, E. S., Shim, J. H., Kang, K. T. Three-dimensional Angiogenesis Assay System using Co-culture Spheroids Formed by Endothelial Colony Forming Cells and Mesenchymal Stem Cells. J. Vis. Exp. (151), e60032, doi:10.3791/60032 (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