Method Article
A reproducible method is presented to generate 3D myocardial tissues combining melt electrospinning writing (MEW) polycaprolactone (PCL) scaffolds and fibrin hydrogels with hiPSC-derived cardiomyocytes and fibroblasts. This technique offers precise control over scaffold architecture and can be applied in preclinical drug testing and cardiac disease modeling.
The development of functional human cardiac tissues holds significant promise for advancing applications in drug screening, disease modeling, and regenerative medicine. This protocol describes the stepwise fabrication of 3D myocardial tissues with advanced mimicry of native cardiac structure by combining melt electrospinning writing (MEW) polycaprolactone (PCL) scaffolds with fibrin hydrogels and human induced pluripotent stem cell (hiPSC)-derived cardiac cells. The process involves embedding a mixture of cardiomyocytes (hiPSC-CMs) and cardiac fibroblasts (hiPSC-CFs) within a fibrin matrix to create mini-tissues, with structural support provided by MEW-generated scaffolds. These fibrillar scaffolds are fabricated at the micro- to nanoscale, allowing for precise control over fiber architecture, which plays a key role in organizing cell distribution and alignment. Meanwhile, the fibrin matrix promotes cell viability and mimics the extracellular environment. Characterization of the generated tissues reveals well-organized sarcomeres within hiPSC-CMs, along with stable contractile activity. The tissues demonstrate consistent spontaneous beating as early as two days post-seeding, with sustained functionality over time. The combination of hiPSC-CFs with hiPSC-CMs enhances the structural integrity of the tissues while supporting long-term cell viability. This approach offers a reproducible, adaptable, and scalable method for creating biomimetic cardiac tissue models, providing a versatile platform for preclinical drug testing, mechanistic studies of cardiac disease, and potential regenerative therapies.
Fabrication of functional human cardiac-engineered tissues holds great promise for high-impact applications. These span from the development of modeling drug cardiotoxicity and human cardiac disease to the generation of therapeutic tissues of relevant sizes1. Although the last 15 years have witnessed significant advances in the field, the fabrication of highly mimetic human myocardium is currently thwarted by difficulties in reproducing the complex structural and mechanical organization of its natural counterpart2.
On the biological side, the revolutionary cell reprogramming technology opened up the possibility of developing patient-specific therapeutic cells. Currently, human induced pluripotent stem cells (hiPSCs) are the only source of human cardiomyocytes in a personalized context. A high yield of cardiomyocytes can be obtained following robust differentiation protocols3,4. A key issue is the degree of maturation as human hiPSC-derived cardiomyocytes (hiPSC-CMs) currently used display an immature phenotype under conventional 2D differentiation conditions5. This is tied to the fact that the structural organization of the cardiac tissue is very complex, absolutely different from conventional 2D cultures. Also, the myocardium is comprised of different cardiovascular cells, including non-myocytes such as cardiac fibroblasts, smooth muscle, and endothelial cells, orderly arranged through a specific 3D structure, allowing efficient blood pumping6,7. Thus, highly structured human cardiac mini tissues composed of all the main cell types are needed to generate physiologically relevant biomimetic tissues.
The application of novel bio-fabrication approaches can break this stalemate. Amongst these, melt electrospinning writing (MEW), an advanced 3D printing technology is able to supply high precision and resolution. In MEW, an electrical field is used to deposit a molten polymer in the form of fibers in the size range of the cell, an order of magnitude smaller than conventional fused deposition modeling (FDM) 3D printing, resulting in highly defined scaffold structures8,9. MEW has been shown capable of translating a complex in silico design into a printed matrix and tuning physical properties in 3D to match those of cardiac tissue10. Using the MEW technology, it is possible to print polymeric meshes with different shapes and structures that, combined with soft cell-friendly hydrogels, generate composite tissues mimicking the micro- and macro-mechanical environment of the native adult myocardium11,12. Modifying the design of the MEW 3D scaffold has been shown to alter the resulting functionality (calcium transients)10, establishing the grounds for understanding the form-function relationship on this promising 3D-engineered system.
Here, a detailed protocol is provided for the fabrication of contractile human cardiac mini tissues from hiPSC-CMs and human iPSC-derived cardiac fibroblasts (hiPSC-CFs) and their combination within a fibrin hydrogel reinforced with 3D MEW printed structures. The addition of fibroblasts has been widely used in different 3D-engineered systems to enhance hiPSC-CMs survival and maintain tissue structure, but its use in 3D-MEW systems has not yet been explored13. The technology described here provides a versatile platform for researchers aiming to develop accurate cardiac tissue models with applications such as understanding disease mechanisms, preclinical toxicology, and drug screening. This model is suited for assessing the cardiotoxicity of established drugs such as anthracyclines (e.g., doxorubicin), tyrosine kinase inhibitors, and emerging therapies like chimeric antigen receptor T (CAR-T) cells, which have been associated with cardiac dysfunction14. Furthermore, the model holds significant potential in advancing regenerative medicine by enabling the testing of biomaterials, bioinks, and cell-based therapies aimed at improving cardiac function and restoring myocardial tissue in conditions such as myocardial infarction.
The details of the reagents and the equipment used in this study are listed in the Table of Materials.
1. Media and reagent setup
2. Human iPSC culture and passage
NOTE: The procedures reported here have been performed with several hiPSC lines, including UCSFi001-A (male, kind gift of Professor Bruce Conklin, David J. Gladstone Institutes), ESi044-A (male), ESi007-A (female) and ESi044-C (female) healthy lines and ESi107-A (cardiac amyloidosis female diseased line) with minor adaptations pertaining hiPSC culture medium, passaging dilution and CHIR concentration. The protocol thereof contains the details specific to the use of UCSFi001-A. All cell culture incubations in this protocol are performed at 37 °C, 5% CO2, and 96% humidity conditions.
3. Human cardiomyocyte differentiation
NOTE: For cardiomyocyte generation from hiPSCs (hiPSC-CMs), the described protocol is based on the monolayer differentiation methodology used by Lian et al.3,15 and Burridge et al.4. With adequate maintenance, hiPSC can be differentiated over 30 consecutive passages with high differentiation efficiency. Signs of abnormal behavior are detected by spontaneous differentiation or consecutive failure of more than 4 differentiations. Regular controls, including mycoplasma testing, are recommended.
4. Human cardiac fibroblast differentiation
NOTE: To obtain human cardiac fibroblasts (hiPSC-CFs) from hiPSCs, the following protocol is based on the two-phase methodology used by Zhang et al.16. The first step is to obtain epicardial cells (hiPSC-EpiCs) by reactivating Wnt signaling pathway after cardiogenic mesoderm induction. Then, hiPSC-EpiCs are exposed to vascular development inhibitors and FGF2 to obtain cardiac fibroblasts in 18 days.
5. Fabrication of Melt Electrospinning Writing (MEW) scaffolds
NOTE: This protocol uses medical grade poly ε-caprolactone (PCL) homopolymer to print fibrillar scaffolds, using a MEW printer specially designed for this purpose by QUT, Queensland University of Technology10.
6. Fibrin mini tissue generation and maintenance
NOTE: The generation of human myocardial 3D mini tissues relies on the encapsulation of hiPSC-derived cardiac cells within fibrin hydrogels combined with MEW scaffolds that provide fibrillary support. The following protocol has been adapted from the engineering design approaches used by Breckwoldt et al.17and Ronaldson-Bouchard et al.18.
7. Systematic analysis of the cardiac differentiation potential of hiPSC and mini tissue function
Characterization of 2D hiPSC-derived cardiac cells
In order to generate multi-phenotypic cardiac tissues, hiPSC-CMs and hiPSC-CFs are independently differentiated and characterized in vitro. With appropriate optimization and strict maintenance, the following protocol will result in a hiPSC-CMs yield of over 80% around day 9 of differentiation, giving rise to spontaneously beating cells (Figure 1A). Furthermore, purity enrichment by metabolic selection largely eliminates non-hiPSC-CMs, resulting in cultures composed of more than 90% of cells expressing cardiac troponin protein (cTNT+) at day 21 (Figure 1B). These robust beating monolayers show expression of the contractile sarcomeric actinin protein (ACTN) by IF staining (Figure 1C, D).
On the other side, the success of this protocol relies on generating cardiac-specific fibroblasts through an intermediate epicardial state induction. Here, after Wnt signaling reactivation by CHIR, epicardial cells are obtained at day 8 from cardiac mesoderm progenitors (Figure 2A). These hiPSC-EpiCs are large cells grouped into single colonies that, when replating with fibroblast medium, give rise to cardiac fibroblast cells with spindle-shaped morphology (Figure 2C). Thus, after 18 total days of differentiation, hiPSC-CFs present over 90% expression of DDR2 by FACS analysis (Figure 2B). This collagen receptor, characterized as a CF marker, is also observable by IF staining (Figure 2D).
Figure 1: hiPSC-CMs differentiation timeline and immunofluorescence and flow cytometry characterization. (A) The overall scheme of the differentiation of CMs derived from hiPSCs and purification steps. (B) Flow cytometry quantification of hiPSC-CMs purity (percentage of cTNT+ cells) at day 21. (C) Representative phase contrast image of cells after purification steps (day 21). Scale bar = 100 µm. (D) Confocal image of fully differentiated hiPSC-CMs stained with ACTN (green) and nuclei stained with DAPI (blue). Scale bar = 25 µm. Please click here to view a larger version of this figure.
Figure 2: hiPSC-CFs differentiation timeline and immunofluorescence and flow cytometry characterization. (A) The overall scheme of the differentiation of CFs derived from hiPSC-epicardial cells. (B) Flow cytometry quantification of hiPSC-CFs purity (percent of DDR2+ cells) at day 18 and representative phase contrast image of the culture (C); scale bar = 100 µm. (D) Confocal image of hiPSC-CFs stained with DDR2 (green) and nuclei stained with DAPI (blue). Scale bar = 25 µm. Please click here to view a larger version of this figure.
Generation of PCL scaffolds by MEW
The mechanical properties of the scaffolds are determined by the total number of layers, fiber density, diameter, distribution, and pore geometry. These are fundamental aspects that can strongly influence cell behavior. Once established, the equipment set up (Figure 3A) and following the printing settings described in this protocol (7 kV, 10 mm collector distance, 2 bar, 80/65 °C head/nozzle, 1080 mm/s collector speed and 23 G syringe tip), 15-layer square scaffolds were generated with square pore geometry of 500 µm x 500 µm (Figure 3B). PCL fibers were deposited layer-by-layer correctly, presenting a fiber diameter of around 15 µm.
Figure 3: MEW equipment and fibrillar scaffold fabrication. (A) MEW setup (i) and Mach3 software (ii); view of the printer head and the collector plate (iii), and Taylor cone formation during printing (iv). (B) Phase contrast images of the fibrillar square scaffolds (i) and amplification of the precise fiber deposition (ii). Scale bar = 250 µm. Please click here to view a larger version of this figure.
Characterization of 3D cardiac mini tissue functionality
Following single-cell dissociation, the Cell Mix composed of 8:2 hiPSC-CMs:hiPSC-CFs was seeded within fibrin hydrogels onto the MEW PCL scaffolds (Figure 4A). After 1 h of incubation at 37 °C, fibrin is stably formed, with cells uniformly distributed throughout the gel, covering the entire pores of the mesh (Figure 4B). This is important since hiPSC-CMs are mostly non-mitotic, and it is not possible to rely on cell growth to fill in the pores as with other cell types. The cells evidenced rapid remodeling of the surrounding matrix by cell elongation, with a localized spontaneous beating as early as 2 days after plating. IF staining analysis showed mixed cell distribution of the cells throughout the gel, interacting with PCL mesh, with a majority of CMs (ACTN+ VIM-) interspaced with VIM+ ACTN- hiPSC-CFs. hiPSC-CMs exhibit a well-organized sarcomere structure marked by regularly spaced ACTN protein staining (Figure 4C). Cardiac tissues of 1 million cells showed stabilized contraction throughout the entire mesh, with a beating frequency of around 30 bpm at day 7 (28.93 ± 11.2, mean ± SD). This functional capacity was sustained throughout all days in culture, with a small decline until day 14 (17.18 ± 12.6, mean ± SD) (Figure 4D). When analyzing metabolic activity, tissues have shown to maintain cell viability throughout culture, with no significant changes, here up analyzed as day 7 vs. day 14 (Figure 4E). Lastly, track point analysis of the tissues at 1 week showed contraction speed of 38.53 µm/s ± 19.8 (mean ± SD, n=16) and contraction amplitude of 28.91 µm ± 15 (mean ± SD, n=16) (Figure 4F).
Figure 4: Generation of human 3D cardiac mini tissues and characterization of tissue structure, functionality, and cell viability. (A) The overall scheme of tissue generation combining hiPSC-derived cardiac cells, MEW printed scaffolds, and fibrin hydrogel encapsulation. (B) Representative phase contrast images of beating mini tissues. Scale bar = 250 µm. (C) Confocal Z-stack image of 3D tissue organization; hiPSC-CMs are stained with ACTN (green), hiPSC-CFs are stained with VIM (red), and nuclei are stained with DAPI (blue). The MEW mesh is indicated by arrowheads. Scale bar = 250 µm. (D) Evolution of beating rate of the tissues from generation to day 14. Data are represented as mean with SD of N = 3, n = 2-4. (E) Cell viability (analyzed by metabolic activity) of tissues between days 7 and 14. Data are represented as mean with SD of N = 3, n = 3-6. (F) Contraction analysis of spontaneously beating cardiac mini tissues at 1 week. Contraction speed (µm/s) and contraction amplitude (µm) are represented as mean with SD (n = 16). Please click here to view a larger version of this figure.
To generate a 3D composite cardiac mini-tissue model that replicates the native characteristics of the human myocardium, several essential factors must be considered. These can be grouped into three main points: (1) optimizing the yield and purity of the cells for tissue fabrication, (2) printing the fibrillar scaffold to mimic the 3D mechano-environment, and (3) integrating the fibrin hydrogel in the fabrication process.
Some of the key steps to ensure a successful differentiation process include maintaining the pluripotency characteristics of the hiPSCs by preventing confluency from exceeding 90%, optimizing the dilution of hiPSC at passaging to achieve adequate confluency at the start of CM or CF differentiation, and fine-tuning the concentration of CHIR for each cell line to ensure efficient mesoderm induction4,15. It is essential to ensure that cultures achieve over 90% purity and that the different cardiac cell types are generated separately. This allows for the formation of functional tissues where the specific roles of each cell type can be properly studied, especially when using them for disease modeling or pharmacological testing. To maintain this high level of cell purity, it is crucial to rigorously carry out the metabolic selection of cardiomyocytes in the glucose-restrictive medium. On the other hand, when obtaining healthy cardiac fibroblasts, the ability to modulate their phenotype and transition into myofibroblasts via the TGF-β pathway offers an opportunity to model conditions like cardiac fibrosis, described elsewhere21. In this protocol, the focus is on generating non-activated, healthy fibroblasts. To achieve this, it is essential to maintain the TGF-β inhibitor (SB) in the culture medium after the differentiation process is completed. Additionally, hiPSC-CFs culture should not exceed six passages, as this could trigger fibroblast activation and transdifferentiation due to the supra-physiological stiffness of tissue culture plastic22.
Moving on to the process of generating artificial 3D scaffolds, MEW stands out as a highly promising technology due to its ability to produce highly ordered micro- to nanoscale fiber architectures using biocompatible materials. Compared to other additive manufacturing technologies, MEW provides superior control over fiber diameter through the adjustment of system parameters such as temperature, collection speed, and applied voltage23,24. However, consistent and precise fiber production requires careful fine-tuning of these parameters to maintain fiber stability. Environmental factors such as room temperature and humidity can introduce minor but significant deviations in scaffold production, underscoring the need for controlled fabrication conditions. Any imbalance can result in alterations in fiber diameter or inaccurate deposition, affecting scaffold morphology and mechanical properties24. Therefore, the optimization of all the above-mentioned parameters is one of the critical steps of this protocol that must be optimized in each laboratory. Here, to fabricate the fibrillar scaffolds integrated into the 3D constructs, PCL medical grade has been selected as the polymer of choice for MEW printing, as it is not only the gold standard in this field but also offers several advantageous material properties. PCL has a low melting point (~60°C) and is semi-conductive, which simplifies the processing requirements and enhances the consistency of fiber formation during the printing. From a biomedical perspective, PCL is highly biocompatible, supporting cell attachment and growth, making it ideal for tissue engineering scaffolds25. Recent innovations also emphasize the use of materials like PCL due to their ability to balance biodegradability with mechanical stability, ensuring that scaffolds provide adequate support for tissue regeneration while gradually degrading without producing harmful byproducts25,26. This is essential for the potential applicability of current constructs for in vivo cardiac regeneration studies. Also, the tunability of MEW scaffold geometries offers further potential for investigating both cell behavior and mechanical properties in tissue-engineered constructs. By modifying pore size and scaffold stiffness, MEW enables the creation of environments that simulate the extracellular matrix influencing cellular responses. This provides a versatile platform for studying cell-scaffold interactions and enhances MEW's potential for advancing the fields of Tissue Engineering and Regenerative medicine26,27,28. In comparison to other techniques like 3D bioprinting, which offers lower precision and resolution, MEW's ability to produce ordered architectures with controlled precision adds tremendous value to its application.
Despite these advantages, MEW presents certain limitations compared to other 3D printing technologies, particularly regarding the maximum height of printable structures. The accumulation of electrostatic charges within the deposited fibers becomes problematic when scaffold heights exceed approximately 4 mm, leading to distortions in the top layers due to charge repulsion29. In this case, the challenge in generating thicker tissues is less about PCL fiber repulsion, as these tissues typically do not exceed 200 µm in thickness, and more about the reduced oxygen availability in thicker constructs (beyond 200-300 µm)30. This reduction in oxygen leads to hypoxia and subsequent cell death. To overcome this issue and make 3D constructs suitable for in vivo regenerative studies, future improvements in tissue thickness will require the integration of vascularization and perfusion. This is crucial, as incorporating vascular networks into 3D constructs will ensure adequate oxygen and nutrient availability throughout the tissue, which is a current focus in the field of cardiac tissue engineering30,31,32.
It is well known that the native myocardium consists of various cardiovascular cells, including cardiomyocytes, fibroblasts, smooth muscle cells, and endothelial cells, all organized in a highly structured and aligned manner7. Although current tissue models incorporate only CMs and CFs, the precise arrangement of these cells, obtained from hiPSCs, within the 3D structure marks a significant advancement. The use of MEW scaffolds plays a crucial role in achieving this organization, as the scaffolds provide a well-defined microstructure that promotes proper alignment and cell organization. In this model, the cardiomyocytes exhibit well-organized sarcomeres and demonstrate strong contraction, which are key characteristics of functional heart tissue. This level of cellular alignment and organization is crucial for mimicking native heart tissue and represents a significant improvement over other models, such as cardiac organoids, where cells often exhibit random alignment33. This model, therefore, provides a more physiologically relevant system for studying heart function and diseases, offering better control over cell organization, which is essential for the accurate modeling of cardiac tissues.
However, to develop truly representative models of heart disease, one significant challenge is overcoming the immaturity of the generated tissues. This issue largely stems from the immature nature of hiPSC-CMs obtained in vitro34. These cells do not fully replicate the functional properties of adult cardiomyocytes, so enhancing their maturation is essential. This can be achieved through two primary approaches: first, by promoting the maturation of the hiPSC-CMs themselves, and second, by improving the maturity of the overall multi-phenotypic tissue. Strategies such as mechanical and electrical stimulation have shown promise in driving both functional and structural maturation, helping to create tissue models that more closely mimic native adult heart tissue35,36. These are applicable to the present model with small changes, given the self-standing nature of the MEW-based tissues.
Regarding the technical steps of the tissue fabrication protocol, it is important to note that the CM: CF ratio embedded in the fibrin hydrogel can vary. It has been reported that for engineered cardiac constructs, a fibroblast proportion of 5%-20% is necessary to mimic the natural beating behavior of the tissues as it could increase the action potential propagation rate37. In this study, a 20% hiPSC-CFs level was selected to better assess fibroblast responses in subsequent toxicology studies. The total number of cells seeded can also be adjusted, as tissues can be regularly generated with 0, 5, 10, or 20% hiPSC-CFs. Always calculate this proportion relative to the total volume of the tissue, not its diameter. Before the process of encapsulation, good cell viability must be ensured. For that, try to discard the TrypLE solution correctly when harvesting cells and avoid keeping the cells disaggregated and pelleted into single cells for too long before seeding. Additionally, as cardiomyocytes are very sensitive to mechanical stress, make sure not to centrifuge at higher g than specified, or pipetting too strongly.
For potential therapeutic applications, it is essential to encapsulate cells within a material that supports both viability and functionality. In this context, fibrin has been chosen due to its high biocompatibility, biodegradability, and ability to mimic components of the extracellular matrix38. The limitations of this material arise from its significant batch-to-batch variability and relatively low mechanical strength. However, these issues are addressed through the reinforcement provided by the MEW fibrillar scaffolds. To ensure the remodeling capacity of the cells within this porous matrix, obtaining a homogeneous solution of the Hydrogel Mix is important. For that, ensure the Cell Mix pellet is sufficiently dry, discard any remaining volume that could dilute the final hydrogel solution, and mix thoroughly with thrombin. For bigger constructs, the Fibrinogen/Thrombin ratio should also be adjusted relative to the total volume of the tissue. Avoiding air bubbles is crucial for achieving a well-defined and homogeneous 3D structure, allowing cells to spread properly. Shortly after the addition of thrombin, an increase in the gel's viscosity should be noticeable, along with a slight color change from pink (Tissue Generation Medium) to yellow.
Lastly, one of the most important steps to ensure the success of the process is adding aprotinin to the medium where the tissues are transferred after the fibrin gel has polymerized for 1 h at 37 °C. Aprotinin, a serine protease inhibitor, prevents fibrinolysis (fibrin degradation) by inhibiting proteolytic enzymes such as plasmin, which are released in the body or culture17,39. Without inhibition, these enzymes would break down the fibrin into its degradation products. By preserving the integrity of the fibrin scaffold, aprotinin allows the matrix to remain functional for extended periods, maintaining the 3D structure essential for supporting long-term cell viability and functionality in the engineered tissues.
The authors have no conflicts of interest.
This research was funded by the H2020 research and innovation program under grant agreements No 874827 (BRAV); Ministerio de Ciencia y Universidades (Spain) through projects PLEC2021-008127 (CARDIOPRINT), PID2022-142562OB-I00 (VOLVAD) and PID2022-142807OA-I00 (INVESTTRA) funded by MICIU/AEI /10.13039/501100011033 and by the European Union NextGenerationEU/PRTR; Gobierno de Navarra Proyectos Estratégicos IMPRIMED (0011-1411-2021-000096) and BIOHEART (0011-1411-2022-000071); Gobierno de Navarra Proyectos Colaborativos BIOGEN (PC020-021-022) and Gobierno de Navarra Salud GN32/2023. Figure 1A, Figure 2A, and Figure 4A are created with BioRender.com.
Name | Company | Catalog Number | Comments |
µ-Slide 8 Well chamber coverslip (IbiTreat) | IBIDI | 80826 | |
0.1% Gelatin Solution (Embryomax) | Merck Millipore | ES-006-B | |
10% formalin | Sigma Aldrich | HT501128 | |
12-well plates | Costar/Corning | 3513 | |
6-well plates | Costar/Corning | 3506 | |
Advanced DMEM 1x (ADMEM) | Gibco | 12491015 | |
Aprotinin from bovine lung | Sigma Aldrich | A1153 | |
B-27 SUPLEMENT, PLUS INSULIN (50x) | Life Technologies | A317504044 | |
B27 SUPPLEMENT, MINUS INSULIN (50x) | Life Technologies | A1895601 | |
Biopsy punch (6mm diameter) | Medical | BP-60F | |
Bovine Serum Albumin (BSA) | Sigma Aldrich | A9647 | |
CHIR-99021 | AXON MEDCHEM | AXON1386 | |
Confocal Laser Scanning Microscope | Zeiss | LSM 800 | |
Culture flask 175 cm | Greiner Bio-One | 660175 | |
Culture flask 75 cm | Falcon | 353136 | |
Cytometer | Beckman Coulter | CytoFlex | |
DAPI | Sigma Aldrich | D9542 | |
Donkey anti-Mouse IgG (H+L) Highly Cross-Adsorbed Secondary Antibody, Alexa Fluor 488 | Invitrogen | A-21202 | |
Donkey anti-Rabbit IgG (H+L) Highly Cross-Adsorbed Secondary Antibody, Alexa Fluor 594 | Invitrogen | A-21207 | |
DPBS (Mg2+, Ca2+ free) | Gibco | A314190094 | |
EDTA 0.5M pH 8.0 | Life Technologies | AM9260G | |
ESSENTIAL 8 MEDIUM KIT | Life Technologies | A1517001 | |
FGF2 (Recombinant Human FGF-basic 154 a.a.) | Peprotech | 100-18B | |
Fibrinogen from bovine plasma | Sigma Aldrich | F8630 | |
Fibroblast Growth Medium 3 KIT (FGM3) | PromoCell | C-23130 | |
Knockout Serum Replacement (KSR) | Life Technologies | 10828028 | |
Lactate (Sodium L-lactate) | Sigma Aldrich | 71718 | |
Matrigel Growth Factor Reduced (MGFR) Basement Membrane Matrix, LDEV-free | Corning | 354230 | |
MEW 23-G needle | Nordson | 7018302 | |
MEW printer | QUT, Queensland University of Technology | ||
MEW syringe | Nordson | 7012072 | |
Mouse Anti-Cardiac Troponin T Monoclonal Antibody | Invitrogen | MA5-12960 | |
Mouse Anti-DDR2 monoclonal antibody | Sigma Aldrich | SAB5300116 | |
Mouse Anti-α-actinin (sarcomeric) Monoclonal Antibody | Sigma Aldrich | A7811 | |
PENICILLIN - STREPTOMYCIN | Life Technologies | 15140122 | |
Poly ε-caprolactone (PCL), medical grade | Corbion | PURASORB® PC 12 | |
Rabbit Anti-Vimentin Recombinant Monoclonal Antibody [EPR3776] - Cytoskeleton Marker | Abcam | Ab29547 | |
Retinoic Acid | Sigma Aldrich | R2625 | |
ROCK inhibitor Y-27632 (10mg) | Fisher Scientific | HB2297 | |
RPMI 1640 (L-glutamine) | Gibco | 21875034 | |
RPMI 1640 (no phenol red) | Gibco | 32404014 | |
RPMI no glucose | Gibco | 11879020 | |
SB431542 | SELLECK CHEMICALS | S1067 | |
Spectrophotometer | BMG Labtech | SPECTROstar Nano | |
Thrombin (human alpha thrombin, Factor IIa) | Enzyme Research Lab | HT 1002a | |
Triton X-100 | Sigma Aldrich | T8787 | |
TrypLE Express | Life Technologies | 12604021 | |
Tween 20 | Sigma Aldrich | P2287 | |
Wnt-C59 | AXON MEDCHEM | AXON2287 |
Request permission to reuse the text or figures of this JoVE article
Request PermissionThis article has been published
Video Coming Soon
Copyright © 2025 MyJoVE Corporation. All rights reserved