Summary

Fabrication and Characterization of Colorectal Cancer Organoids from SW1222 Cell Line in Ultrashort Self-Assembling Peptide Matrix

Published: May 03, 2024
doi:

Summary

This protocol aims to evaluate biofunctional self-assembling peptides for cell adhesion, organoid morphology, and gene expression by immunostaining. We will use a colorectal cancer cell line to provide a cost-effective way of obtaining organoids for intensive testing.

Abstract

Ultrashort self-assembling peptides (SAPs) can spontaneously form nanofibers that resemble the extracellular matrix. These fibers allow the formation of hydrogels that are biocompatible, biodegradable, and non-immunogenic. We have previously proven that SAPs, when biofunctionalized with protein-derived motifs, can mimic the extracellular matrix characteristics that support colorectal organoid formation. These biofunctional peptide hydrogels retain the original parent peptide’s mechanical properties, tunability, and printability while incorporating cues that allow cell-matrix interactions to increase cell adhesion. This paper presents the protocols needed to evaluate and characterize the effects of various biofunctional peptide hydrogels on cell adhesion and lumen formation using an adenocarcinoma cancer cell line able to form colorectal cancer organoids cost-effectively. These protocols will help evaluate biofunctional peptide hydrogel effects on cell adhesion and luminal formation using immunostaining and fluorescence image analysis. The cell line used in this study has been previously utilized for generating organoids in animal-derived matrices.

Introduction

In recent years, self-assembling peptides (SAPs) have emerged as promising biomaterials for tissue engineering applications. SAPs possess unique properties, including spontaneous formation of nanofibers, biocompatibility, biodegradability, and non-immunogenicity, making them attractive candidates for scaffold development1. SAPs have been previously used together with various types of cells, and notably, reported ultrashort SAPs have facilitated the encapsulation of stem cells while upholding their pluripotency over prolonged periods encompassing more than 30 passages and with minimal incidence of chromosomal aberrations2,3,4. Hence, adapting SAPs for their use in organoid culture constituted a rational subsequent step.

Organoids are complex three-dimensional structures that arise from single pluripotent cells. These structures give rise to various cell types, which then self-organize to replicate the developmental processes of embryonic and tissue growth in vitro5. This innovative framework has evolved into a formidable tool for in vitro investigations, efficiently conserving the genetic, phenotypic, and behavioral attributes characteristic of in vivo organs6. Nevertheless, a principal impediment in the bench-to-bedside transition of organoid-based findings is the need for reproducibility7. This variation in results is primarily ascribed to the difficulty in fully differentiating human pluripotent stem cells into specialized cell lineages, compounded by the absence of authentic tissue architecture and the inherent complexity encountered in organoid models. Given the rise in popularity of stem-cell- and organoid-based studies8, there has been an escalated demand for biomaterials with dynamic mechanical properties and integrin-like adhesion sites or scaffolds with controlled degradability7,9. These attributes can be effectively tuned through the strategic utilization of biofunctionalized SAPs.

SAPs are short sequences of amino acids with the inherent ability to spontaneously organize into well-defined structures10. These peptides often contain alternating hydrophobic and hydrophilic residues, which drive their self-assembly through non-covalent interactions, including hydrogen bonding, electrostatic interactions, and hydrophobic effects11,12. The self-assembly process of these peptides is primarily driven by the need to minimize the system's free energy. When in an aqueous environment, the hydrophobic residues tend to cluster together to minimize their exposure to water, while the hydrophilic residues interact with the surrounding water molecules. This phenomenon leads to the formation of various nanostructures. In this case, ultrashort self-assembling peptides form nanofibers with characteristics that depend on the peptide sequence and environmental conditions2,12,13,14,15,16. These peptides adopt a β-turn conformation, where individual peptide strands align parallel or antiparallel to each other, stabilized by hydrogen bonds (Figure 1). The presence of positive ions accelerates the self-assembling processes that lead to nanofiber formation.

Peptide nanofibers have been identified as possessing an innate ability to entrap significant volumes of water. This characteristic facilitates the generation of a hydrogel, which subsequently manifests as a biocompatible and biodegradable three-dimensional space conducive to cellular proliferation and growth. These self-assembling peptide nanofibers intricately emulate the topographical features inherent to the natural extracellular matrix (ECM) environment1. This resemblance endows cells with an environment that imitates their physiological habitat, further promoting optimal cellular activities17. Furthermore, the versatility inherent to these peptides permits a considerable degree of tunability4. Such adaptability enables changes to the matrix's properties, such as stiffness, gelation kinetics, and porosity. These adaptations are achieved through modifications to the peptide sequence. Consequently, self-assembling peptides (SAPs) have emerged as pivotal components in contemporary biomaterial science, extensively used as scaffolds for tissue regeneration and cellular culture13,17.

One of the critical advantages of self-assembling peptides is their ability to be easily synthesized and modified at the molecular level. This advantage allows for the incorporation of specific functional groups or bioactive motifs into the peptide sequence, enabling the design of peptides with tailored properties and functionalities18,19,20 (Figure 1B). For example, biofunctionalized SAPs can be designed to mimic the ECM and promote differentiation using the RGD peptide19,21. Peptide Ben-IKVAV has also been reported to significantly increase the expression of neuronal-specific markers due to its ligand-specific motiety22. These peptides can also be engineered to display bioactive molecules, such as integrin-binding peptides, to enhance cell survival23. Finally, other biofunctionalized SAPs have been developed to promote angiogenesis by including the IKVAV and YIGSR motifs in their structures24.

Biofunctionalized SAPs reported in an earlier publication show that self-assembling can be further modified by including the naïve peptide from which the biofunctionalized SAP was derived25. These SAP mixtures can also provide a wide array of SAP formulations that vary in their biochemical activity and physicochemical properties. For instance, the amphiphilic peptide IIFK is an ultrashort SAP that can withstand biofunctionalization with various motifs, exemplified by the incorporation of the IKVAV motif (Figure 1B). Both peptides can form nanofibers, both alone and combined. Each formulation results in hydrogels with varying physical properties (Figure 2)

However, because of the extensive array of potential permutations and alternatives obtained from the biofunctionalization of SAPs, there is a need to provide a fast and cost-effective option for organoid testing. One candidate for such intensive and cost-effective organoid evaluation is the SW1222 cell line, derived from human colorectal adenocarcinoma26,27. SW1222 cells possess characteristics that enable their aggregation into 3D structures resembling organoids, making them an ideal model for studying tissue development and regenerative medicine applications. SW1222 cells have been identified as capable of generating organoids owing to their intrinsic overexpression of the LGR5 gene27. The creation of organoids from individual LGR5+ stem cells has been convincingly exhibited before, as well as the propensity of SW1222 cells to achieve morphological characteristics of a colorectal cancer organoid28.

In this methods paper, we present detailed protocols for evaluating and characterizing the effects of various biofunctional peptides on cell adhesion and lumen formation using SW1222 cells (Figure 3). By providing step-by-step procedures and imaging analysis methods, we hope to offer valuable insights into the biofabrication of organoids and the evaluation of simplistic SAP matrices for organoid culture.

Protocol

1. Buffer and solution preparation NOTE: All stated concentrations are final concentrations. Add fetal bovine serum (FBS) and penicillin-streptomycin at a final concentration of 10% and 1%, respectively, to prepare complete Iscove's Modified Dulbecco's Medium (IMDM). Store in the dark at 4 °C for up to 1 month. Mix MgCl2 (3 mM), sucrose (300 mM), and Triton X-100 (0.5%) in phosphate buffer saline (PBS) to prepare the permeabilizati…

Representative Results

First, we evaluated the cells grown in a 24-well plate for 7 days using brightfield imaging. We identified small clusters of cells assembling into organoids during the week, as seen in Figure 4. A controlled scan method can follow the mobility of the cells and the organoids between different days. In general, we looked at the evolution of the morphology of the cells during the whole week. SW1222-derived organoids should have a round morphology and a light appearance. A darker appearance indi…

Discussion

The vast potential of SAPs in biomedical research is underscored by their malleability and adaptability through biofunctionalization. With such an extensive array of permutations, the challenge arises in efficiently testing and determining the most promising configurations for specific applications, especially in organoid research. A fast, cost-effective solution is paramount. The SW1222 cell line, derived from human colorectal adenocarcinoma, emerges as a pivotal candidate for such intensive evaluation. Here, we present…

Disclosures

The authors have nothing to disclose.

Acknowledgements

This work was financially supported by King Abdullah University of Science and Technology. The authors acknowledge KAUST's Seed Fund Grant and KAUST's Innovation Fund awarded by KAUST's Innovation and Economic Development. The authors would like to acknowledge KAUST's Bioscience and Imaging Core Labs for supporting the biological characterization and microscopy analyses.

Materials

1x PBS Gibco 14190144
6-well plate, tissue culture treated Corning 07-200-83
10x PBS, no calcium, no magnesium Gibco 70011044
16% Formaldehyde (w/v), Methanol-free Thermo Scientific 28906
24-well plate, tissue culture treated Corning 09-761-146
96-well black plate, tissue culture treated Corning 07-200-565
alamarBlue Cell Viability Reagent Invitrogen DAL1025
Anti-Ezrin antibody, rabbit monoclonal Abcam ab40839 Secondary used: anti-rabbit Dylight 633
Anti-pan Cadherin antibody, rabbit polyclonal Abcam ab16505 Secondary used: anti-rabbit Alexa 488
Anti-ZO1 tight junction antibody, goat polyclonal Abcam ab190085 Secondary used: anti-goat Alexa 488
BSA Sigma-Aldrich A9418
Cellpose 2.0 NA NA Obtained from https://github.com/MouseLand/cellpose
Confocal Laser Scanning Microscope with Airyscan ZEISS LSM 880
DAPI Invitrogen D1306
Donkey anti-Goat IgG (H+L) Secondary Antibody, Alexa Fluor 488 Invitrogen A-11055
Glycine Cytiva GE17-1323-01
Goat anti-Rabbit IgG (H+L) Secondary Antibody, Alexa Fluor 488 Invitrogen A-11008
Goat anti-Rabbit IgG (H+L) Secondary Antibody, DyLight 633 Invitrogen 35562
Heat Inactivated Fetal Bovine Serum (HI FBS) Gibco 16140071
ImageJ 1.54f NIH NA
IMDM Gibco 12440079
LIVE/DEAD Viability/Cytotoxicity Kit, for mammalian cells Invitrogen L3224
Magnesium Chloride, hexahydrate (MgCl2 6 H2O) Sigma-Aldrich M2393
Matrigel for Organoid Culture, phenol-red free Corning 356255 Refered in the manuscript as Matrigel or basement membrane matrix.
Microscope, brightfield
Microscope, EVOS Thermo Scientific EVOS M7000
OriginPro 2023 (64-bit) 10.0.0.154 OriginLab Corp NA
Penicillin-Streptomycin (10,000 U/mL) Gibco 15140122
Peptide P (Ac,Ile,Ile,Phe,Lys,NH2) Lab-made NA Can be custom-made by peptide manufacturers such as Bachem.
Peptide P1 (Ac, Ile, Ile, Phe, Lys, Gly, Gly, Gly, Arg, Gly, Asp, Ser, NH2) Lab-made NA Can be custom-made by peptide manufacturers such as Bachem.
Peptide P2 (Ac, Ile,Ile,Phe,Lys,Gly,Gly,Gly,Ile,Lys,Val,Ala,Val,NH2) Lab-made NA Can be custom-made by peptide manufacturers such as Bachem.
Rhodamine Phalloidin Invitrogen R415
Round Cover Slip, 10 mm diameter VWR 631-0170
Scanning Electron Microscope Thermo Fisher – FEI TENEO VS
Sterile 30 μm strainer Sysmex 04-004-2326
sucrose Sigma-Aldrich S1888
SW1222 cell line ECACC 12022910
Triton x-100 Thermo Scientific 85111
Trypsin-EDTA 0.25% Gibco 25200056
Tween 20 Sigma-Aldrich P1379
UltraPure water Invitrogen 10977015

References

  1. Susapto, H. H., et al. Ultrashort peptide bioinks support automated printing of large-scale constructs assuring long-term survival of printed tissue constructs. Nano Letters. 21 (7), 2719-2729 (2021).
  2. Loo, Y., et al. A chemically well-defined, self-assembling 3D substrate for long-term culture of human pluripotent stem cells. ACS Applied Bio Materials. 2 (4), 1406-1412 (2019).
  3. Loo, Y., et al. Self-assembled proteins and peptides as scaffolds for tissue regeneration. Advanced Healthcare Materials. 4 (16), 2557-2586 (2015).
  4. Loo, Y., et al. Peptide bioink: self-assembling nanofibrous scaffolds for three-dimensional organotypic cultures. Nano Letters. 15 (10), 6919-6925 (2015).
  5. Ho, B. X., Pek, N. M. Q., Soh, B. -. S. Disease modeling using 3D organoids derived from human induced pluripotent stem cells. International Journal of Molecular Sciences. 19 (4), 936 (2018).
  6. Li, Y., Tang, P., Cai, S., Peng, J., Hua, G. Organoid based personalized medicine: from bench to bedside. Cell Regeneration. 9 (1), 21 (2020).
  7. Kim, S., et al. Tissue extracellular matrix hydrogels as alternatives to Matrigel for culturing gastrointestinal organoids. Nature Communications. 13 (1), 1692 (2022).
  8. Kaushik, G., Ponnusamy, M. P., Batra, S. K. Concise review: current status of three-dimensional organoids as preclinical models. Stem Cells. 36 (9), 1329-1340 (2018).
  9. Gjorevski, N., et al. Designer matrices for intestinal stem cell and organoid culture. Nature. 539 (7630), 560-564 (2016).
  10. Hauser, C. A., et al. Natural tri- to hexapeptides self-assemble in water to amyloid beta-type fiber aggregates by unexpected alpha-helical intermediate structures. Proceedings of the National Academy of Sciences of the United States of America. 108 (4), 1361-1366 (2011).
  11. Lakshmanan, A., Zhang, S., Hauser, C. A. E. Short self-assembling peptides as building blocks for modern nanodevices. Trends in Biotechnology. 30 (3), 155-165 (2012).
  12. Rauf, S., et al. Self-assembling tetrameric peptides allow in situ 3D bioprinting under physiological conditions. Journal of Materials Chemistry B. 9 (4), 1069-1081 (2021).
  13. Bilalis, P., et al. Dipeptide-based photoreactive instant glue for environmental and biomedical applications. ACS Applied Materials & Interfaces. 15 (40), 46710-46720 (2023).
  14. Abdelrahman, S., et al. The impact of mechanical cues on the metabolomic and transcriptomic profiles of human dermal fibroblasts cultured in ultrashort self-assembling peptide 3D scaffolds. ACS Nano. 17 (15), 14508-14531 (2023).
  15. Moretti, M., et al. Selectively positioned catechol moiety supports ultrashort self-assembling peptide hydrogel adhesion for coral restoration. Langmuir. 39 (49), 17903-17920 (2023).
  16. Alhattab, D. M., et al. Fabrication of a three-dimensional bone marrow niche-like acute myeloid Leukemia disease model by an automated and controlled process using a robotic multicellular bioprinting system. Biomaterials Research. 27 (1), 111 (2023).
  17. Abdelrahman, S., et al. The impact of mechanical cues on the metabolomic and transcriptomic profiles of human dermal fibroblasts cultured in ultrashort self-assembling peptide 3D scaffolds. ACS Nano. 17 (15), 14508-14531 (2023).
  18. Habibi, N., Kamaly, N., Memić, A., Shafiee, H. Self-assembled peptide-based nanostructures: smart nanomaterials toward targeted drug delivery. Nano Today. 11 (1), 41-60 (2016).
  19. Yan, Y., et al. Enhanced osteogenesis of bone marrow-derived mesenchymal stem cells by a functionalized silk fibroin hydrogel for bone defect repair. Advanced Healthcare Materials. 8 (3), 1801043 (2018).
  20. Kisiday, J., et al. Self-assembling peptide hydrogel fosters chondrocyte extracellular matrix production and cell division: implications for cartilage tissue repair. Proceedings of the National Academy of Sciences of the United States of America. 99 (15), 9996-10001 (2002).
  21. Hogrebe, N. J., et al. Independent control of matrix adhesiveness and stiffness within a 3D self-assembling peptide hydrogel. Acta Biomaterialia. 70, 110-119 (2018).
  22. Wu, F. J., Lin, H. C. Synthesis, self-assembly, and cell responses of aromatic IKVAV peptide amphiphiles. Molecules. 27 (13), 4115 (2022).
  23. Cringoli, M. C., et al. Bioadhesive supramolecular hydrogel from unprotected, short d,l-peptides with Phe-Phe and Leu-Asp-Val motifs. Chemical Communications. 56 (20), 3015-3018 (2020).
  24. Pulat, G. O., Gökmen, O., Çevik, Z. B. Y., Karaman, O. Role of functionalized self-assembled peptide hydrogels in in vitro vasculogenesis. Soft Matter. 17 (27), 6616-6626 (2021).
  25. Pérez-Pedroza, R., et al. Fabrication of lumen-forming colorectal cancer organoids using a newly designed laminin-derived bioink. International Journal of Bioprinting. 9 (1), 633 (2022).
  26. Ashley, N., Yeung, T. M., Bodmer, W. F. Stem cell differentiation and lumen formation in colorectal cancer cell lines and primary tumors. 암 연구학. 73 (18), 5798-5809 (2013).
  27. Yeung, T. M., Gandhi, S. C., Wilding, J. L., Muschel, R., Bodmer, W. F. Cancer stem cells from colorectal cancer-derived cell lines. Proceedings of the National Academy of Sciences of the United States of America. 107 (8), 3722-3727 (2010).
  28. Sato, T., et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature. 459 (7244), 262-265 (2009).
  29. Pachitariu, M., Stringer, C. Cellpose 2.0: how to train your own model. Nature Methods. 19 (12), 1634-1641 (2022).
  30. Stringer, C., Wang, T., Michaelos, M., Pachitariu, M. Cellpose: a generalist algorithm for cellular segmentation. Nature Methods. 18 (1), 100-106 (2021).
  31. Adan, A., Kiraz, Y., Baran, Y. Cell proliferation and cytotoxicity assays. Current Pharmaceutical Biotechnology. 17 (14), 1213-1221 (2016).
  32. Humphries, M. J. Cell-substrate adhesion assays. Current Protocols in Cell Biology. , (2001).
  33. Munevar, S., Wang, Y. -. l., Dembo, M. Distinct roles of frontal and rear cell-substrate adhesions in fibroblast migration. Molecular Biology of the Cell. 12 (12), 3947-3954 (2001).
  34. Djomehri, S., Burman, B., González, M. E., Takayama, S., Kleer, C. G. A reproducible scaffold-free 3D organoid model to study neoplastic progression in breast cancer. Journal of Cell Communication and Signaling. 13 (1), 129-143 (2018).
  35. Puschhof, J., et al. Intestinal organoid cocultures with microbes. Nature Protocols. 16 (10), 4633-4649 (2021).
  36. Matthews, J. M., et al. OrganoID: A versatile deep learning platform for tracking and analysis of single-organoid dynamics. PLOS Computational Biology. 18 (11), 1010584 (2022).
  37. Herpers, B., et al. Functional patient-derived organoid screenings identify MCLA-158 as a therapeutic EGFR × LGR5 bispecific antibody with efficacy in epithelial tumors. Nature Cancer. 3 (4), 418-436 (2022).
  38. Borten, M. A., Bajikar, S. S., Sasaki, N., Clevers, H., Janes, K. A. Automated brightfield morphometry of 3D organoid populations by OrganoSeg. Scientific Reports. 8 (1), 5319 (2018).
  39. Bergdorf, K. N., et al. Immunofluorescent staining of cancer spheroids and fine-needle aspiration-derived organoids. STAR Protocols. 2 (2), 100578-100578 (2021).
  40. Yang, Z., Xu, H., Zhao, X. Designer self-assembling peptide hydrogels to engineer 3D cell microenvironments for cell constructs formation and precise oncology remodeling in ovarian cancer. Advanced Science. 7 (9), 1903718 (2020).
  41. Yadav, N., et al. Ultrashort peptide-based hydrogel for the healing of critical bone defects in rabbits. ACS Applied Materials & Interfaces. 14 (48), 54111-54126 (2022).
  42. Alshehri, S., Susapto, H. H., Hauser, C. A. E. Scaffolds from self-assembling tetrapeptides support 3D spreading, osteogenic differentiation, and angiogenesis of mesenchymal stem cells. Biomacromolecules. 22 (5), 2094-2106 (2021).
This article has been published
Video Coming Soon
Keep me updated:

.

Cite This Article
Perez-Pedroza, R., Moretti, M., Hauser, C. A. E. Fabrication and Characterization of Colorectal Cancer Organoids from SW1222 Cell Line in Ultrashort Self-Assembling Peptide Matrix. J. Vis. Exp. (207), e66060, doi:10.3791/66060 (2024).

View Video