Summary

Comparative Study of Basement-Membrane Matrices for Human Stem Cell Maintenance and Intestinal Organoid Generation

Published: March 15, 2024
doi:

Summary

Organoids have become valuable tools for disease modeling. The extracellular matrix (ECM) guides cell fate during organoid generation, and using a system that resembles the native tissue can improve model accuracy. This study compares the generation of induced pluripotent stem cells-derived human intestinal organoids in animal-derived ECM and xeno-free hydrogels.

Abstract

Extracellular matrix (ECM) plays a critical role in cell behavior and development. Organoids generated from human induced pluripotent stem cells (hiPSCs) are in the spotlight of many research areas. However, the lack of physiological cues in classical cell culture materials hinders efficient iPSC differentiation. Incorporating commercially available ECM into stem cell culture provides physical and chemical cues beneficial for cell maintenance. Animal-derived commercially available basement membrane products are composed of ECM proteins and growth factors that support cell maintenance. Since the ECM holds tissue-specific properties that can modulate cell fate, xeno-free matrices are used to stream up translation to clinical studies. While commercially available matrices are widely used in hiPSC and organoid work, the equivalency of these matrices has not been evaluated yet. Here, a comparative study of hiPSC maintenance and human intestinal organoids (hIO) generation in four different matrices: Matrigel (Matrix 1-AB), Geltrex (Matrix 2-AB), Cultrex (Matrix 3-AB), and VitroGel (Matrix 4-XF) was conducted. Although the colonies lacked a perfectly round shape, there was minimal spontaneous differentiation, with over 85% of the cells expressing the stem cell marker SSEA-4. Matrix 4-XF led to the formation of 3D round clumps. Also, increasing the concentration of supplement and growth factors in the media used to make the Matrix 4-XF hydrogel solution improved hiPSC expression of SSEA-4 by 1.3-fold. Differentiation of Matrix 2-AB -maintained hiPSC led to fewer spheroid releases during the mid-/hindgut stage compared to the other animal-derived basement membranes. Compared to others, the xeno-free organoid matrix (Matrix 4-O3) leads to larger and more mature hIO, suggesting that the physical properties of xeno-free hydrogels can be harnessed to optimize organoid generation. Altogether, the results suggest that variations in the composition of different matrices affect stages of IO differentiation. This study raises awareness about the differences in commercially available matrices and provides a guide for matrix optimization during iPSC and IO work.

Introduction

The extracellular matrix (ECM) is a dynamic and multifunctional component of tissues that plays a central role in regulating cell behavior and development. As a complex network, it provides structural support, cell adhesive ligands1, and storage of growth factors and cytokines that regulate cell signaling. For example, during wound healing, the ECM serves as a scaffold for migrating cells and as a reservoir of growth factors involved in tissue repair2. Similarly, dysregulation in the ECM can lead to an increase in the severity of various diseases such as fibrosis and cancer3,4. During embryonic development, the ECM guides tissue morphogenesis. For example, in the development of the heart, ECM components play a role in creating the correct architecture and function of the heart tissue5. Over a decade of research has shown that the stiffness of the microenvironment alone6,7 can control stem cell lineage specification. Therefore, it is not surprising that during in vitro cell differentiation, ECM influences stem cell fate by providing signals for differentiation.

Organoids can be generated from induced pluripotent stem cells (iPSCs). Starting with a properly characterized iPSC line is required to generate organoids successfully. However, the lack of physiological cues in classical cell culture materials hinders efficient iPSC differentiation and organoid generation. Moreover, recent research has emphasized the significance of the composition of the extracellular matrix (ECM), interactions between cells and the ECM8, as well as mechanical and geometrical cues9,10,11 in the context of organoid expansion and differentiation12. Advancing organoid technology by improving reproducibility will involve incorporating tissue-specific physical and chemical cues.

Organoids aim to recapitulate the native tissue within a physiologically similar microenvironment. Choosing an ECM system that closely mimics the native tissue ECM is crucial for achieving physiological relevance regarding cell behavior, function, and response to stimuli13. The choice of ECM components can influence the differentiation of stem cells into specific cell types within the organoid. Different ECM proteins and their combinations can provide cues that guide cell fate14. For example, studies have shown that using specific ECM components can promote the differentiation of intestinal stem cells into mature intestinal cell types, resulting in physiologically relevant intestinal organoids15. While organoids are a valuable tool during disease modeling and drug testing, selecting an appropriate ECM system is pivotal to this application. An appropriate ECM system can enhance the accuracy of disease modeling by creating a microenvironment that resembles the affected tissue16. Furthermore, tissue-specific ECM can help generate organoids that better recapitulate disease-associated phenotypes and drug responses17. Optimizing the ECM system used in organoid differentiation is critical for achieving desired differentiation outcomes.

Commercially available basement membrane systems derived from animal ECM sources (e.g., Matrigel, Cultrex) and xeno-free hydrogel (e.g., VitroGel) are widely used in iPSC and organoid research. Companies that commercialize them and researchers that use them have laid out many instructions for their specific products and applications over the years. Many of these instructions served as a guide for the generation of this protocol. Furthermore, the benefits and setbacks associated with their intrinsic properties have been individually noted by many18,19,20,21. However, there is no systematic workflow to guide the selection of optimal systems for iPSC and organoid work. Here, a workflow to systematically evaluate the equivalency of ECM systems from various sources for iPSC and organoid work is provided. This is a comparative study of the maintenance of two different human iPSC lines (hiPSC) and human intestinal organoids (hIO) generation in four different matrices: Matrigel (Matrix 1-AB), Geltrex (Matrix 2-AB), Cultrex (Matrix 3-AB), and VitroGel (Matrix 4-XF). For organoid culture, four versions of the xeno-free matrix VitroGel that were previously optimized for organoid culture were used: ORGANOID 1 (Matrix 4-O1), ORGANOID 2 (Matrix 4-O2), ORGANOID 3 (Matrix 4-O3), ORGANOID 4 (Matrix 4-O4). Also, animal-derived matrices optimized for organoids were used: Matrigel High Concentration (Matrix 1-ABO) and Cultrex Type 2 (Matrix 3-ABO). Commercially available stem cell culture media (mTeSR Plus) and organoid differentiation kit (STEMdiff intestinal organoid kit) were used. This protocol combines the individual instructions from the products' manufacturers with lab experiences to guide the reader toward a successful optimization of ECM for their specific iPSC and organoid work. Altogether, this protocol and representative results emphasize the importance of selecting the optimal microenvironment for stem cell work and organoid differentiation.

Protocol

1. hiPSC maintenance CAUTION: All work is done in a Biosafety Cabinet (BSC) following standard aseptic techniques. Must follow OSHA safety standards for laboratories, including proper use of personal protective equipment such as lab coats, gloves, and goggles. Preparation of matrices aliquots and cell culture media For commercially available animal-derived basement membranes (BMs; Matrix 1-AB, Matrix 2-AB, Matrix 3-AB), prepare aliquots of working volum…

Representative Results

Following this protocol, commercially available basement membranes and a xeno-free hydrogel system were successfully utilized to cultivate hiPSC cells and differentiate them into hIO. The main objective of these experiments was to systematically evaluate the equivalency of matrices from various sources for hiPSC and hIO work. The first section of this protocol focused on the maintenance and characterization of a healthy iPSC culture that yields an efficient intestinal organoid generation. The process of coating the cultu…

Discussion

Selecting the optimal microenvironment for stem cell and organoid work is a pivotal early step when using these platforms for a wide range of applications. Our representative results show that Matrix 4-XFO3, in combination with a higher concentration of growth factors, leads to larger organoids, suggesting that the physical properties of xeno-free hydrogels can be harnessed to optimize organoid generation using these systems. It has been previously shown that the unique characteristics of the extracellular matrix (ECM) a…

Offenlegungen

The authors have nothing to disclose.

Acknowledgements

The authors acknowledge previous training and general recommendations regarding starting hiPSC and organoid work from Drs. Christina Pacak, Silveli Susuki-Hatano, and Russell D'Souza. They thank Dr. Chelsey Simmons for her guidance in using hydrogel systems for in vitro cell culture work. Also, the authors would like to thank Drs. Christine Rodriguez and Thomas Allison from STEMCELL Technologies for their guidance on hiPSC culture. The authors also thank TheWell Bioscience for covering the publication costs.

Materials

24-Well Plate (Culture treated, sterile) Falcon 353504
37 °C water bath VWR
96-well plate  Fisher Scientific FB012931
Advanced DMEM/F12 Life Technologies 12634
Anti-adherence Rinsing Solutio STEMCELL Technologies 7010
Biological safety cabinet (BSC) Labconco  Logic
Brightfield Microscope Echo Rebel REB-01-E2
BXS0116 ATCC ACS-1030
Centrifuge with temperature control (4 °C capabilities) ThermoScientific 75002441
Conical tubes, 15 mL, sterile Thermo Fisher Scientific 339650
Conical tubes, 50 mL, sterile Thermo Fisher Scientific 339652
Cultrex RGF BME, Type 2 Bio-techne 3533-005-02
Cultrex Stem Cell Qualified RGF BME  Bio-techne 3434-010-02
D-PBS (Without Ca++ and Mg++) Thermo Fisher Scientific 14190144
GeltrexLDEV-Free, hESC-Qualified Reduce Growth Factor Gibco  A14133-02
GlutaMAX Supplement Thermo Fischer Scientific 35050-061
Guava Muse Cell Analyzer or another flow cytometry equipment (optional) Luminex 0500-3115
HEPES buffer solution Thermo Fischer Scientific 15630-056
Heralcell Vios Cell culture incubator (37 °C, 5% CO2) Thermo Scientific  51033775
JMP Software SAS Institute JMP 16
MATLAB MathWorks, Inc R2022b
Matrigel Growth Factor Reduced (GFR) Basement Membrane Matrix LDEV free Corning  356231
Matrigel Matrix High Concentration (HC), Growth Factor Reduced (GFR) LDEV-free Corning  354263
mTeSR Plus Medium STEMCELL Technologies 100-0276
Nunclon Delta surface treated 24-well plate Thermo Scientific 144530
PE Mouse Anti-human CD326 (EpCAM) BD Pharmingen 566841
PE Mouse Anti-human CDX2  BD Pharmingen 563428
PE Mouse Anti-human FOXA2 BD Pharmingen 561589
PerCP-Cy 5.5 Mouse Anti-human SSEA4  BD Pharmingen 561565
ReLeSR STEMCELL 5872
SCTi003-A STEMCELL Technologies 200-0510
Serological pipettes (10 mL)  Fisher Scientific 13-678-11E
Serological pipettes (5 mL)  Fisher Scientific 13-678-11D
STEMdiff Intestinal Organoid Growth Medium STEMCELL Technologies 5145
STEMdiff Intestinal Organoid Kit STEMCELL Technologies 5140
Vitrogel Hydrogel Matrix TheWell Bioscience VHM01
VitroGel ORGANOID Discovery Kit TheWell Bioscience VHM04-K

Referenzen

  1. Hynes, R. O. Integrins: Bidirectional, allosteric signaling machines. Cell. 110 (6), 673-687 (2002).
  2. Frantz, C., Stewart, K. M., Weaver, V. M. The extracellular matrix at a glance. J Cell Sci. 123, 4195-4200 (2010).
  3. Hinz, B., Gabbiani, G. Fibrosis: Recent advances in myofibroblast biology and new therapeutic perspectives. F1000 Biol Rep. 2, 78 (2010).
  4. Pickup, M. W., Mouw, J. K., Weaver, V. M. The extracellular matrix modulates the hallmarks of cancer. EMBO Rep. 15 (12), 1243-1253 (2014).
  5. Rozario, T., DeSimone, D. W. The extracellular matrix in development and morphogenesis: A dynamic view. Dev Biol. 341 (1), 126-140 (2010).
  6. Even-Ram, S., Artym, V., Yamada, K. M. Matrix control of stem cell fate. Cell. 126 (4), 645-647 (2006).
  7. Engler, A. J., Sen, S., Sweeney, H. L., Discher, D. E. Matrix elasticity directs stem cell lineage specification. Cell. 126 (4), 677-689 (2006).
  8. Tran, O. N., et al. Organ-specific extracellular matrix directs trans-differentiation of mesenchymal stem cells and formation of salivary gland-like organoids in vivo. Stem Cell Res Ther. 13 (1), 306 (2022).
  9. Nikolaev, M., et al. Homeostatic mini-intestines through scaffold-guided organoid morphogenesis. Nature. 585 (7826), 574-578 (2020).
  10. Gjorevski, N., et al. Designer matrices for intestinal stem cell and organoid culture. Nature. 539 (7630), 560-564 (2016).
  11. Gjorevski, N., et al. Tissue geometry drives deterministic organoid patterning. Science. 375 (6576), (2022).
  12. Heo, J. H., Kang, D., Seo, S. J., Jin, Y. Engineering the extracellular matrix for organoid culture. Int J Stem Cells. 15 (1), 60-69 (2022).
  13. Shamir, E. R., Ewald, A. J. Three-dimensional organotypic culture: experimental models of mammalian biology and disease. Nat Rev Mol Cell Biol. 15 (10), 647-664 (2014).
  14. Clevers, H. Modeling development and disease with organoids. Cell. 165 (7), 1586-1597 (2016).
  15. Jung, P., et al. Isolation and in vitro expansion of human colonic stem cells. Nat Med. 17 (10), 1225-1227 (2011).
  16. Lancaster, M. A., Knoblich, J. A. Organogenesis in a dish: Modeling development and disease using organoid technologies. Science. 345 (6194), 1247125 (2014).
  17. Huch, M., et al. In vitro expansion of single Lgr5+ liver stem cells induced by Wnt-driven regeneration. Nature. 494 (7436), 247-250 (2013).
  18. Greenlee, A. R., Kronenwetter-Koepel, T. A., Kaiser, S. J., Liu, K. Comparison of Matrigel and gelatin substrata for feeder-free culture of undifferentiated mouse embryonic stem cells for toxicity testing. Toxicol In Vitro. 19 (3), 389-397 (2005).
  19. Geltrex LDEV-Free, HESC-Qualified, Reduced Growth Factor Basement Membrane Matrix User Guide (Pub.No. MAN0007336 3.0. Fisher Scientific Available from: https://www.thermofisher.com/document-connect/document-connect.html?url=https://assets.thermofisher.cn/TFS-Assets%2FLSG%2Fmanuals%2FGeltrex_LDEV_Free_hESC_qualified_PI.pdf (2024)
  20. biotechne R&D Systems. Cultrex Stem Cell Qualified Reduced Growth Factor. biotechne R&D Systems. , (2024).
  21. VitroGel Organoid Protocol. TheWell Bioscience Available from: https://www.thewellbio.com/video-protocols (2024)
  22. Spence, J. R., et al. Directed differentiation of human pluripotent stem cells into intestinal tissue in vitro. Nature. 470 (7332), 105-110 (2011).
  23. Henderson, J. K., et al. Preimplantation human embryos and embryonic stem cells show comparable expression of stage-specific embryonic antigens. Stem Cells. 20 (4), 329-337 (2002).
  24. Haruna, N. F., Huang, J. Investigating the dynamic biophysical properties of a tunable hydrogel for 3D cell culture. J Cytol Tissue Biol. 7, 30 (2020).
  25. Cherne, M. D., et al. A synthetic hydrogel, VitroGel ORGANOID-3, improves immune cell-epithelial interactions in a tissue chip co-culture model of human gastric organoids and dendritic cells. Front Pharmacol. 12, 707891 (2021).
  26. Stewart, D. C., et al. Quantitative assessment of intestinal stiffness and associations with fibrosis in human inflammatory bowel disease. PLoS One. 13, e0200377 (2018).
  27. Hernandez-Gordillo, V., et al. Fully synthetic matrices for in vitro culture of primary human intestinal enteroids and endometrial organoids. Biomaterials. 254, 120125 (2020).
  28. Broguiere, N., et al. Growth of epithelial organoids in a defined hydrogel. Adv Mater. 30, 1801621 (2018).
  29. Barthes, J., et al. Cell microenvironment engineering and monitoring for tissue engineering and regenerative medicine: The recent advances. BioMed Res Int. 2014, 921905 (2014).
  30. Engler, A. J., Sen, S., Sweeney, H. L., Discher, D. E. Matrix elasticity directs stem cell lineage specification. Cell. 126 (4), 677-689 (2006).
  31. Aisenbrey, E. A., Murphy, W. L. Synthetic alternatives to Matrigel. Nat Rev Mater. 5 (7), 539-551 (2020).

Play Video

Diesen Artikel zitieren
Pineiro-Llanes, J., da Silva, L., Huang, J., Cristofoletti, R. Comparative Study of Basement-Membrane Matrices for Human Stem Cell Maintenance and Intestinal Organoid Generation. J. Vis. Exp. (205), e66277, doi:10.3791/66277 (2024).

View Video