JoVE Logo
Faculty Resource Center

Sign In

Summary

Abstract

Introduction

Protocol

Representative Results

Discussion

Acknowledgements

Materials

References

Bioengineering

Controlled Strain of 3D Hydrogels under Live Microscopy Imaging

Published: December 4th, 2020

DOI:

10.3791/61671

1Department of Biomedical Engineering, Faculty of Engineering, Tel-Aviv University, 2Department of Materials Science and Engineering, Faculty of Engineering, Tel-Aviv University, 3School of Mechanical Engineering, Faculty of Engineering, Tel-Aviv University, 4Center for the Physics and Chemistry of Living Systems, Tel-Aviv University

The presented method involves uniaxial stretching of 3D soft hydrogels embedded in silicone rubber while allowing live confocal microscopy. Characterization of the external and internal hydrogel strains as well as fiber alignment are demonstrated. The device and protocol developed can assess the response of cells to various strain regimes.

External forces are an important factor in tissue formation, development, and maintenance. The effects of these forces are often studied using specialized in vitro stretching methods. Various available systems use 2D substrate-based stretchers, while the accessibility of 3D techniques to strain soft hydrogels, is more restricted. Here, we describe a method that allows external stretching of soft hydrogels from their circumference, using an elastic silicone strip as the sample carrier. The stretching system utilized in this protocol is constructed from 3D-printed parts and low-cost electronics, making it simple and easy to replicate in other labs. The experimental process begins with polymerizing thick (>100 μm) soft fibrin hydrogels (Elastic Modulus of ~100 Pa) in a cut-out at the center of a silicone strip. Silicone-gel constructs are then attached to the printed-stretching device and placed on the confocal microscope stage. Under live microscopy the stretching device is activated, and the gels are imaged at various stretch magnitudes. Image processing is then used to quantify the resulting gel deformations, demonstrating relatively homogenous strains and fiber alignment throughout the gel’s 3D thickness (Z-axis). Advantages of this method include the ability to strain extremely soft hydrogels in 3D while executing in situ microscopy, and the freedom to manipulate the geometry and size of the sample according to the user’s needs. Additionally, with proper adaptation, this method can be used to stretch other types of hydrogels (e.g., collagen, polyacrylamide or polyethylene glycol) and can allow for analysis of cells and tissue response to external forces under more biomimetic 3D conditions.

Tissue response to mechanical forces is an integral part of a wide range of biological functions, including gene expression1, cell differentiation2, and tissue remodeling3. Moreover, force-induced changes in the extracellular matrix (ECM) such as fiber alignment and densification can impact cell behavior and tissue formation4,5,6. The ECM’s fibrous mesh structure has intriguing mechanical properties, such as non-linear elasticity, non-affine deformation and plastic deformations7

Log in or to access full content. Learn more about your institution’s access to JoVE content here

1. Solution preparation (to be performed in advance)

  1. Fibrinogen labeling
    NOTE: The labeling step is required only if analyzing the deformation of the fibrin gel is desired. For cellular experiments, it is possible to use an unlabeled gel.
    1. Add 38 μL of 10 mg/mL succinimidyl ester fluorescent dye (dissolved in DMSO) to 1.5 mL of 15 mg/mL fibrinogen solution (molar ratio of 5:1) in a 50 mL centrifuge tube and place on a shaker for 1 hour at room temperature. Afterwards, place the tube in .......

Log in or to access full content. Learn more about your institution’s access to JoVE content here

Representative data from static stretch of increasing magnitudes applied to the silicone strip carrying a 3D fibrin hydrogel, embedded with 1 μm fluorescent beads, is shown in Figure 9. The analysis demonstrates the effect of silicone stretch on geometric changes of the cut-out as well as the developed strains within the gel. Z-stack images of the entire gel are used to evaluate the deformation of the original circle shaped cut-out to the elliptical geometry (

Log in or to access full content. Learn more about your institution’s access to JoVE content here

The method and protocol presented herein are largely based on our previous study by Roitblat Riba et al.41 We include here the full computer-aided design (CAD), Python and microcontroller codes of the SCyUS device.

The major advantages of the presented method over existing approaches include the possibility to strain very soft 3D hydrogels (Elastic Modulus of ~100 Pa) from their circumference, and under live confocal imaging. Other methods are usually .......

Log in or to access full content. Learn more about your institution’s access to JoVE content here

Some figures included here have been adapted by permission from the Copyright Clearance Center: Springer Nature, Annals of Biomedical Engineering. Straining 3D hydrogels with uniform z-axis strains while enabling live microscopy imaging, A. Roitblat Riba, S. Natan, A. Kolel, H. Rushkin, O. Tchaicheeyan, A. Lesman, Copyright© (2019).

https://doi.org/10.1007/s10439-019-02426-7

....

Log in or to access full content. Learn more about your institution’s access to JoVE content here

NameCompanyCatalog NumberComments
Alexa Fluor 546 carboxylic acid, succinimidyl esterInvitrogenA20002
Cell Medium (DMEM High Glucose)Biological Industries01-052-1AAdd 10% FBS, 1% PNS, 1% L-Glutamine, 1% Sodium Pyruvate
Cover Slip #1.5Bar-Naor Ltd.BN72204-3022×40 mm
DIMETHYL SULPHOXIDE 99.5% GC DMSOSigma-Aldrich Inc.D-5879-500 ML
Dulbecco's Phosphate-Buffered SalineBiological Industries02-023-1A
EVICEL Fibrin Sealant (Human)Omrix Biopharmaceuticals3902Fibrinogen: 70 mg/mL, Thrombin: 800-1200 IU/mL
Fibrinogen BufferN/ARecipe for 1L: 7g NaCl, 2.94g trisodium citrate dihydrate, 9g glycine, 20g arginine hydrochloride & 0.15g calcium chloride dihydrate. Bring final volume to 1L with PuW (pH 7.0-7.2)
Fluorescent micro-beads FluoSpheres (1 µm)InvitrogenF8820Orange (540/560)
Provided as suspension (2% solids) in water plus 2 mM sodium azide
High-Temperature Silicone RubberMcMaster-Carr3788T41580 µm-thick
E = 1.5 Mpa
Poisson Ratio = 0.48
Tensile Strength = 4.8 MPa
Upper limit of stretch = +300% engineering strain
HiTrap desalting column 5 mL (Sephadex G-25 packed)GE Healthcare17-1408-01
HIVAC-G High Vacuum Sealing CompoundShin-Etsu Chemical Co., Ltd.HIVAC-G 100
ImageJ FIJI software39National Institute of Health, Bethesda, MDVersion 1.8.0_112
Microcontroller (Adruino Uno + Adafruit Motorshield v2.3)Arduino/AdafruitArduino-DK001/Adafruit-1438
MicroVL 21R CentrifugeThermo Scientific75002470
ParafilmBemisPM-996
Primovert Light MicroscopeCarl Zeiss Suzhou Co., Ltd.491206-0011-000
SCyUS CAD (Solidworks)Dassault SystèmesN/A
SCyUS Code37N/AN/A
Servomotor - TowerPro SG-5010Adafruit155
SL 16R CentrifugeThermo Scientific75004030For 50 mL tubes
Sterile 10 cm non-culture platesCorning430167
Thrombin bufferN/ARecipe for 1L: 20g mannitol, 8.77g NaCl, 2.72g sodium acetate trihydrate, 24 mL 25% Human Serum Albumin, 5.88g calcium chloride. Bring final volume to 1L with PuW (pH 7.0)
Trypsin EDTA Solution B (0.25%), EDTA (0.05%)Biological Industries03-052-1B
USB Cable (Type B Male to Type A Male)N/AN/A
Zeiss LSM 880 Confocal MicroscopeCarl Zeiss AG2811000417
ZEN 2.3 SP1 FP3 (black)Carl Zeiss AGRelease Version 14.0.0.0

  1. Bleuel, J., Zaucke, V., Bruggemann, G. P., Niehoff, A. Effects of cyclic tensile strain on chondrocyte metabolism: a systematic review. PLoS ONE. 10, 0119816 (2015).
  2. Pennisi, C. P., Olesen, C. G., de Zee, M., Rasmussen, J., Zachar, V. Uniaxial cyclic strain drives assembly and differentiation of skeletal myocytes. Tissue Engineering Part A. 17, 2543-2550 (2011).
  3. Grodzinsky, A. J., Levenston, M. E., Jin, M., Frank, E. H. Cartilage Tissue Remodeling in Response to Mechanical Forces. Annual Review of Biomedical Engineering. 2 (1), 691-713 (2000).
  4. Munster, S., et al. Strain history dependence of the nonlinear stress response of fibrin and collagen networks. Proceedings of the National Academy of Sciences of the USA. 110, 12197-12202 (2013).
  5. Vader, D., Kabla, A., Weitz, D., Mahadevan, L. Strain-induced alignment in collagen gels. PLoS ONE. 4, 5902 (2009).
  6. Badylak, S. F. The extracellular matrix as a scaffold for tissue reconstruction. Seminars in Cell & Developmental Biology. 13 (5), 377-383 (2002).
  7. Natan, S., Koren, Y., Shelah, O., Goren, S., Lesman, A. . Molecular Biology of the Cell. 31 (14), 1474-1485 (2020).
  8. Ban, E., et al. Mechanisms of Plastic Deformation in Collagen Networks Induced by Cellular Forces. Biophysical Journal. 114 (2), 450-461 (2018).
  9. Kim, J., et al. Stress-induced plasticity of dynamic collagen networks. Nature Communications. 8, 842 (2017).
  10. Storm, C., Pastore, J. J., MacKintosh, F. C., Lubensky, T. C., Janmey, P. A. Nonlinear elasticity in biological gels. Nature. 435, 191-194 (2005).
  11. Wen, Q., Basu, A., Janmey, P. A., Yodh, A. G. Non-affine deformations in polymer hydrogels. Soft Matter. 8, 8039-8049 (2012).
  12. Muiznieks, L. D., Keeley, F. W. Molecular assembly and mechanical properties of the extracellular matrix: A fibrous protein perspective. Biochimica et Biophysica Acta. 1832, 866-875 (2012).
  13. Brown, A. E. X., Litvinov, R. I., Discher, D. E., Purohit, P. K., Weisel, J. W. Multiscale mechanics of fibrin polymer: gel stretching with protein unfolding and loss of water. Science. 325, 741-744 (2009).
  14. Carroll, S. F., Buckley, C. T., Kelly, D. J. Cyclic tensile strain can play a role in directing both intramembranous and endochondral ossification of mesenchymal stem cells. Frontiers in Bioengineering and Biotechnology. 5, 73 (2017).
  15. Livne, A., Bouchbinder, E., Geiger, B. Cell reorientation under cyclic stretching. Nature Communications. 5, 3938 (2014).
  16. Wang, L., et al. Patterning cellular alignment through stretching hydrogels with programmable strain gradients. ACS Applied Materials & Interfaces. 7, 15088-15097 (2015).
  17. Xu, G. K., Feng, X. Q., Gao, H. Orientations of Cells on Compliant Substrates under Biaxial Stretches: A Theoretical Study. Biophysical Journal. 114 (3), 701-710 (2017).
  18. Chagnon-Lessard, S., Jean-Ruel, H., Godin, M., Pelling, A. E. Cellular orientation is guided by strain gradients. Integrative Biology (United Kingdom). 9 (7), 607-618 (2013).
  19. Lu, J., et al. Cell orientation gradients on an inverse opal substrate. ACS Applied Materials & Interfaces. 7 (19), 10091-10095 (2015).
  20. Baker, B. M., Chen, C. S. Deconstructing the third dimension - 3D culture microenvironments alter cellular cues. Journal of Cell Science. 125, 3015-3024 (2012).
  21. Bono, N., et al. Unraveling the role of mechanical stimulation on smooth muscle cells: a comparative study between 2D and 3D models. Biotechnology and Bioengineering. 113, 2254-2263 (2016).
  22. Pampaloni, F., Reynaud, E. G., Stelzer, E. H. K. The third dimension bridges the gap between cell culture and live tissue. Nature Reviews Molecular Cell Biology. 8, 839-845 (2007).
  23. Riehl, B. D., Park, J. H., Kwon, I. K., Lim, J. Y. Mechanical stretching for tissue engineering: two-dimensional and three-dimensional constructs. Tissue Engineering Part B: Reviews. 18, 288-300 (2012).
  24. Flexcell. Linear Tissue Train Culture Plate. Flexcell. , (2019).
  25. Flexcell. Tissue Train. Flexcell. , (2019).
  26. CellScale. MCT6 Stretcher. CellScale. , (2019).
  27. STREX. STB-150. STREX. , (2019).
  28. STREX. Stretch Chambers. STREX. , (2019).
  29. Kamble, H., Barton, M. J., Jun, M., Park, S., Nguyen, N. T. Cell stretching devices as research tools: engineering and biological considerations. Lab on a Chip. 16, 3193-3203 (2016).
  30. Weidenhamer, N. K., Tranquillo, R. T. Influence of cyclic mechanical stretch and tissue constraints on cellular and collagen alignment in fibroblast-derived cell sheets. Tissue Engineering Part C: Methods. 19, 386-395 (2013).
  31. Yung, Y. C., Vandenburgh, H., Mooney, D. J. Cellular strain assessment tool (CSAT): precision-controlled cyclic uniaxial tensile loading. Journal of Biomechanics. 42, 178-182 (2009).
  32. Chen, K., et al. Role of boundary conditions in determining cell alignment in response to stretch. Proceedings of the National Academy of Sciences of the USA. 115, 986-991 (2018).
  33. Heher, P., et al. A novel bioreactor for the generation of highly aligned 3D skeletal muscle-like constructs through orientation of fibrin via application of static strain. Acta Biomaterialia. 24, 251-265 (2015).
  34. Foolen, J., Deshpande, V. S., Kanters, F. M. W., Baaijens, F. P. T. The influence of matrix integrity on stress-fiber remodeling in 3D. Biomaterials. 33, 7508-7518 (2012).
  35. Walker, M., Godin, M., Pelling, A. E. A vacuum-actuated microtissue stretcher for long-term exposure to oscillatory strain within a 3D matrix. Biomedical Microdevices. 20, 43 (2018).
  36. Zhao, R. G., Boudou, T., Wang, W. G., Chen, C. S., Reich, D. H. Decoupling cell and matrix mechanics in engineered microtissues using magnetically actuated microcantilevers. Advanced Materials. 25, 1699-1705 (2013).
  37. Li, Y. H., et al. Magnetically actuated cell-laden micro-scale hydrogels for probing strain-induced cell responses in three dimensions. NPG Asia Materials. 8, 238 (2016).
  38. Li, Y. H., et al. An approach to quantifying 3D responses of cells to extreme strain. Scientific Reports. 6, 19550 (2016).
  39. Humphrey, J. D., et al. A theoretically-motivated biaxial tissue culture system with intravital microscopy. Biomechanics and Modeling in Mechanobiology. 7, 323-334 (2008).
  40. Niklason, L. E., et al. Enabling tools for engineering collagenous tissues integrating bioreactors, intravital imaging, and biomechanical modeling. Proceedings of the National Academy of Sciences of the USA. 107, 3335-3339 (2010).
  41. Roitblat Riba, A., et al. Straining 3D hydrogels with uniform z-axis strains while enabling live microscopy imaging. Annals of Biomedical Engineering. , (2019).
  42. Gomez, D., Natan, S., Shokef, Y., Lesman, A. Mechanical interaction between cells facilitates molecular transport. Advanced Biosystems. 3 (12), 1900192 (2019).
  43. Schindelin, J., et al. Fiji: an open- source platform for biological-image analysis. Nature Methods. 9, 676-682 (2012).
  44. EPFL Switzerland. OrientationJ plug in. EPFL Switzerland. , (2019).
  45. Goren, S., Koren, Y., Xu, X., Lesman, A. Elastic anisotropy governs the decay of cell-induced displacements. Biophysical Journal. 118 (5), 1152-1164 (2019).
  46. Notbohm, J., Lesman, A., Tirrell, D. A., Ravichandran, G. Quantifying cell-induced matrix deformation in three dimensions based on imaging matrix fibers. Integrative Biology. 7 (10), 1186-1195 (2015).
  47. Lesman, A., Notbohm, J., Tirrell, D. A., Ravichandran, G. Contractile forces regulate cell division in three-dimensional environments. Journal of Cell Biology. 205 (2), 155-162 (2014).
  48. Cha, C. Y., et al. Tailoring Hydrogel Adhesion to Polydimethylsiloxane Substrates Using Polysaccharide Glue. Angewandte Chemie International Edition. 52, 6949-6952 (2019).
  49. Wirthl, D., et al. Instant tough bonding of hydrogels for soft machines and electronics. Science Advances. 3, (2017).
  50. Juarez-Moreno, J. A., Avila-Ortega, A., Oliva, A. I., Aviles, F., Cauich-Rodriguez, J. V. Effect of wettability and surface roughness on the adhesion properties of collagen on PDMS films treated by capacitively coupled oxygen plasma. Applied Surface Science. 349, 763-773 (2015).
  51. Kim, H. T., Jeong, O. C. PDMS surface modification using atmospheric pressure plasma. Microelectronic Engineering. 88, 2281-2285 (2011).
  52. Prasad, B. R., et al. Controlling cellular activity by manipulating silicone surface roughness. Colloids and Surfaces. 78, 237-242 (2010).

This article has been published

Video Coming Soon

JoVE Logo

Privacy

Terms of Use

Policies

Research

Education

ABOUT JoVE

Copyright © 2024 MyJoVE Corporation. All rights reserved