Summary

Flow Cytometry and Confocal Imaging Analysis of Low Wnt Expression in Axin2-mTurquoise2 Reporter Thymocytes

Published: September 24, 2021
doi:

Summary

Signaling levels are known to regulate cell fate, indicating that regulation of Wnt signaling constitutes an interesting therapeutic target. Here, we describe flow cytometry and confocal microscopy analysis methods for a robust murine canonical Wnt signaling reporter model that measures distinct Wnt signaling levels.

Abstract

Measuring Wnt expression levels is essential when trying to identify or test new Wnt therapeutic targets. Previous studies have shown that canonical Wnt signaling operates via a dosage-driven mechanism, motivating the need to study and measure Wnt signaling in various cell types. Although several reporter models have been proposed to represent physiological Wnt expression, either the genetic context or the reporter protein highly influenced the validity, accuracy, and flexibility of these tools. This paper describes methods for acquiring and analyzing data obtained with the Axin2-mTurquoise2 mouse Wnt reporter model, which contains a mutated Axin2em1Fstl allele. This model facilitates the study of endogenous canonical Wnt signaling in individual cells over a wide range of Wnt activity.

This protocol describes how to fully appreciate Axin2-mTurquoise2 reporter activity using cell population analysis of the hematopoietic system, combined with cell surface markers or β-catenin intracellular staining. These procedures serve as a base for implementation and reproduction in other tissues or cells of interest. By combining fluorescence-activated cell sorting and confocal imaging, distinct canonical Wnt expression levels can be visualized. The recommended measurement and analysis strategies provide quantitative data on the fluorescent expression levels for precise assessment of canonical Wnt signaling. These methods will be useful for researchers who want to use the Axin2-mTurquise2 model for canonical Wnt expression patterns.

Introduction

Canonical Wnt signaling is a conserved signaling pathway implicated in healthy tissue homeostasis as well as in disease. Precise regulation of Wnt signaling levels has been shown to be important in embryonic development, but is also of great importance in adult tissues. Canonical Wnt signaling has been found to play an important role in tissue regeneration of several organs such as the gut, the skin, and the hematopoietic system. Hence, when Wnt signaling is deregulated, severe pathologies arise. Colorectal, liver, and skin cancer, neurological disease, as well as certain hematological malignancies are exemplary pathologies wherein deregulated Wnt signaling is the causative factor or contributor1. Therefore, several inhibitors for different Wnt targets are currently being tested in clinical trials as Wnt-associated cancer therapeutics2.

Additionally, interesting advances are taking place in Wnt therapeutic potential for neurological recovery, age-related neurological disorders, and congenital autism spectrum disorders3,4,5. Wnt signals have been explored for ex vivo expansion of stem cells for subsequent transplantation6. However, therapeutic targeting of canonical Wnt signaling is a difficult endeavor due to its importance in many basic cell functions and cross-talk with other pathways7,8,9, resulting in the need to precisely measure the effects of these Wnt therapeutic agents in an easy-to-interpret model. Canonical Wnt signaling is driven by short-range, soluble Wnt ligands, which are secreted by neighboring cells or as autocrine excretion as reported in various Wnt-responsive stem cell types.

The Wnt Frizzled receptor and lipoprotein receptor-related protein (LRP) co-receptors are responsive to these ligands, which triggers an intracellular signaling cascade. When Wnt signaling is off, a destruction complex composed of Axis Inhibitor (Axin), tumor suppressor gene product, Adenomatous Polyposis Coli (APC), Casein Kinase1 (CK1α), and Glycogen Synthase Kinase (GSK-3β), prevents the accumulation of β-catenin (CTNNB1) by proteasomal degradation. Upon Wnt ligand-receptor binding, the destruction complex is inactivated, leading to accumulation and stabilization of β-catenin in the cytoplasm. The active β-catenin can migrate to the nucleus where it binds to the Transcription Factor/Lymphoid Enhancer-binding Factor (TCF/LEF) transcription factors to initiate the transcription of Wnt target genes. Axin2 is considered a target gene as it is a direct target of the Wnt pathway10. Additionally, Axin2 serves as a negative regulator as well as a reporter gene for active canonical Wnt signaling11,12.

Several canonical Wnt signaling reporters have been described in literature and have been of great use in understanding the role of Wnt signaling in embryonic development. Most of these reporters make use of synthetically inserted TCF/LEF binding sites, which do not use an endogenous target gene13,14,15,16,17,18,19. Additionally, Axin2 knock-in strategies have been used that respect the natural location of the gene11,20,21,22,23, of which Axin2-LacZ is generally accepted as the most robust canonical Wnt reporter11. However, the reporter protein LacZ, albeit easy-to-use in most tissues, requires a β-galactosidase substrate, which is recognized to be harsh for live cells24. Especially for stem cells and thymocytes, the harsh LacZ detection conditions increase cellular death (own unreported data) when handling cell suspensions.

Although the signal amplification caused by the LacZ staining is convenient to detect low signals, it makes the quantification less direct and thus arguably less reliable. Therefore, a murine reporter model was designed to mimic the Axin2-LacZ genetic strategy, but with an mTurquoise2 reporter protein21, to provide a readout that is more direct and closer to the physiological expression levels. The mTurquoise2 fluorescent protein is an excellent substitute for LacZ due to its high brightness (quantum yield (QY)= 0.93), flexibility in combination with other fluorescent proteins for extensive cell surface characterization, and its lack of needing an exogenous substrate. Furthermore, its close genetic relationship to green fluorescent protein (GFP) offers the possibility to use most GFP-recognizing fluorescent antibodies for stronger signal detection, if necessary, in extremely Wnt-sensitive cells25.

The Axin2-mTurquoise2 model is not only a canonical Wnt reporter, but also offers the possibility to study Axin2 heterozygote and homozygote (Axin2 knock-out) phenotypes. The targeted insertion of mTurquoise2 at the start site of Axin2 results in a disrupted Axin2 protein21. As Axin2, also known as Conductin, is part of the Wnt destruction complex, and the destruction complex tightly regulates β-catenin mediated transcription, its partial or complete absence could be of interest to study diverse pathologies. For instance, in colorectal cancer, Axin2 levels are relatively high due to Wnt hyperactivation11; however, its role in other pathologies is still largely unknown. Even though Axin2 is considered to play a limited role in the degradation of β-catenin, its role in Wnt regulation can be enhanced by the addition of a small peptide, which blocks Wnt-mediated colorectal cancer growth26.

Altogether, careful Wnt regulation via Wnt therapeutic targets can open up opportunities to change the onset or development of severe pathologies and should be further investigated in models with reporter capacity. In this report, we explain our best-practice analysis method of the Axin2-mTurquoise2 murine model for flow cytometry and confocal imaging. In the context of Wnt dosage levels, very low canonical Wnt signaling levels are difficult to detect, for which advanced detection and analysis abilities provide an advantage to fully derive the benefits of this model. Thymocytes are used as a model system due to their fragile cell viability, low canonical Wnt signaling expression, and condensed cytoplasm area to represent the detection sensitivity of the Axin2-mTurquoise2 model. Additionally, a histological total β-catenin-staining procedure for thymocyte cell suspensions is explained to measure cytoplasmic β-catenin levels and verify nuclear active canonical Wnt signaling in combination with the reporter.

Protocol

NOTE: All mouse procedures were performed with the approval of the Leiden University Medical Centre (LUMC) Ethical Committee on Animal Experiments. Male and female, 6-12-week-old, wild-type (wt), which have no insertion of the Axin2-mTurquoise2 reporter construct, heterozygous (Tg/0) with one insertion of the Axin2-murquoise2 reporter construct and thus, one disrupted Axin2 gene, and homozygous (Tg/Tg) with the insertion of the Axin2-mTurquoise2 reporter construct in both alleles and thus, two …

Representative Results

To investigate the role of canonical Wnt signaling, an Axin2-mTurquoise2 canonical Wnt reporter model has been tested in combination with β-catenin protein expression. Thymocytes are known to be fragile, show low canonical Wnt signaling at several stages in the thymocyte maturation process, and have a low cytoplasmic to nuclear ratio; all these factors hinder the detection of cytoplasmic mTurquoise2 or β-catenin. By following the protocol, murine Axin2-mTurquoise2 thymocytes were harvested from the thymus and p…

Discussion

Several canonical Wnt reporters are available with differing reporter sensitivity and actual reporter proteins. Reporter models using synthetically introduced multimerized TCF/LEF binding sites are available with fluorescent reporter proteins; however, such repeats of transgenes can be lost during breeding or long in vivo experiments and can be sensitive to non-Wnt signals from surrounding genomic sequences that influence reporter expression. Therefore, the most used reporter remains the older variant Axin2-LacZ…

Disclosures

The authors have nothing to disclose.

Acknowledgements

This work was supported in part by a grant from Leiden University for the profiling Area Regenerative Medicine to develop novel mouse models.

Materials

BD FACScantoII flow cytometer BD Biosciences not aplicable Serial number V96300710. The flow cytometer setup in this protocol contains a 405 nm laser line with 505 longpass filter and 530/30 nm bandpass filter, and 470/20 nm bandpass filter; a 488 nm laser with 735 nm longpass filter and 780/60 nm bandpass filter, 670 nm longpass filter and 655 nm longpass filter, 610 nm longpass filter, 550 nm longpass filter and 575/26 nm bandpass filter, 505 nm longpass filter and 530/30 nm bandpass filter, and 488/10 nm bandpass filter; and a 633 nm laser line with 735 nm longpass filter and 780/60 nm bandpass filter, 685 nm longpass filter, and 660/20 nm bandpass filter.
BSA  Sigma  A9647
Corning 70 μm cell strainer  Falcon/Corning  352350
Cytospin 4 Type A78300101 Thermo Scientific not aplicable 
DMSO Sigma Aldrich  D5879-1L
DNAse I Sigma  A9647
Falcon 50 mL Conical Centrifuge tubes Greiner bio-one 227261
Falcon round-bottom Polystyrene Test tubes with cell strainer snap cap Fisher Scientific  352235
Fetal Calf Serum (FCS) Greiner Bio-One B.V.  not aplicable  Depends on origin
Fiji software  ImageJ not aplicable  Version 1.53
Filter card white (for cytospin) VWR SHAN5991022
FlowJo 10 software Treestar not aplicable  Version 10.5.3
Frost slides  Klinipath
Gibco IMDM medium  Fisher Scientific  12440053
HCX PL APLO 40x 1.4 OIL lens Leica microsystems  not aplicable 
Hydrophobic pen: Omm Edge pen  Vector not aplicable 
Leica TCS SP5 DMI6000 Leica microsystems  not aplicable  The microscope setup in this protocol consisted of an HCX PL APO 40x/1.2 oil-immersion objective with 8-bit resolution, 1024 pixels x 1024 pixels, 400 Hz speed, pinhole 68 µm, and zoom factor of 1.5 at room temperature. This system contains a 405 diode laser, argon laser, DPSS 561 laser, HeNe 594 laser and HeNe 633 laser with 4 hybrid detectors (HyDs) and 5 photomultiplier tubes (PMTs). 
Methanol  VWR 1060091000
NaN3/sodium azide Hospital farmacy not aplicable 
Normal mouse serum Own mice not aplicable 
PBS  Lonza BE17-517Q
ProLong Diamond Antifade Mountant Fisher Scientific  P36965
Purified mouse anti-β-catenin (CTNNB1) BD Biosciences 610154
TO-PRO-3 Iodide Thermofisher T3605
Transparent nailpolish at any drugstore not aplicable 
Tween-20 Sigma Aldrich  P1379-500ml 
Zenon Alexa Fluor 568 Mouse IgG1 labeling kit Thermofisher Z25006

References

  1. Kahn, M. Can we safely target the WNT pathway. Nature Reviews. Drug Discovery. 13 (7), 513-532 (2014).
  2. Jung, Y. S., Park, J. I. Wnt signaling in cancer: therapeutic targeting of Wnt signaling beyond beta-catenin and the destruction complex. Experimental & Molecular Medicine. 52 (2), 183-191 (2020).
  3. Gao, K., Zhang, T., Wang, F., Lv, C. Therapeutic Potential of Wnt-3a in neurological recovery after spinal cord injury. European Neurology. 81 (3-4), 197-204 (2019).
  4. Jia, L., Pina-Crespo, J., Li, Y. Restoring Wnt/beta-catenin signaling is a promising therapeutic strategy for Alzheimer’s disease. Molecular Brain. 12 (1), 104 (2019).
  5. Bae, S. M., Hong, J. Y. The Wnt signaling pathway and related therapeutic drugs in autism spectrum disorder. Clinical Psychopharmacology and Neuroscience. 16 (2), 129-135 (2018).
  6. Tajer, P., Pike-Overzet, K., Arias, S., Havenga, M., Staal, F. J. T. Ex vivo expansion of hematopoietic stem cells for therapeutic purposes: Lessons from development and the niche. Cells. 8 (2), 169 (2019).
  7. Yanai, K., et al. Crosstalk of hedgehog and Wnt pathways in gastric cancer. Cancer Letters. 263 (1), 145-156 (2008).
  8. Blank, U., et al. An in vivo reporter of BMP signaling in organogenesis reveals targets in the developing kidney. BMC Developmental Biology. 8, 86 (2008).
  9. Duncan, A. W., et al. Integration of Notch and Wnt signaling in hematopoietic stem cell maintenance. Nature Immunology. 6 (3), 314-322 (2005).
  10. Jho, E. H., et al. Wnt/beta-catenin/Tcf signaling induces the transcription of Axin2, a negative regulator of the signaling pathway. Molecular and Cellular Biology. 22 (4), 1172-1183 (2002).
  11. Lustig, B., et al. Negative feedback loop of Wnt signaling through upregulation of conductin/Axin2 in colorectal and liver tumors. Molecular and Cellular Biology. 22 (4), 1184-1193 (2002).
  12. Bernkopf, D. B., Hadjihannas, M. V., Behrens, J. Negative-feedback regulation of the Wnt pathway by conductin/axin2 involves insensitivity to upstream signalling. Journal of Cell Science. 128 (1), 33-39 (2015).
  13. Vassar, R., Rosenberg, M., Ross, S., Tyner, A., Fuchs, E. Tissue-specific and differentiation-specific expression of a human K14 keratin gene in transgenic mice. Proceedings of the National Academy of Sciences of the United States of America. 86 (5), 1563-1567 (1989).
  14. DasGupta, R., Fuchs, E. Multiple roles for activated LEF/TCF transcription complexes during hair follicle development and differentiation. Development. 126 (20), 4557-4568 (1999).
  15. Maretto, S., et al. Mapping Wnt/beta-catenin signaling during mouse development and in colorectal tumors. Proceedings of the National Academy of Sciences of the United States of America. 100 (6), 3299-3304 (2003).
  16. Mohamed, O. A., Clarke, H. J., Dufort, D. beta-catenin signaling marks the prospective site of primitive streak formation in the mouse embryo. Developmental Dynamics. 231 (2), 416-424 (2004).
  17. Moriyama, A., et al. GFP transgenic mice reveal active canonical Wnt signal in neonatal brain and in adult liver and spleen. Genesis. 45 (2), 90-100 (2007).
  18. Currier, N., et al. Dynamic expression of a LEF-EGFP Wnt reporter in mouse development and cancer. Genesis. 48 (3), 183-194 (2010).
  19. Ferrer-Vaquer, A., et al. A sensitive and bright single-cell resolution live imaging reporter of Wnt/beta-catenin signaling in the mouse. BMC Developmental Biology. 10, 121 (2010).
  20. Jho, E. H., et al. Wnt/beta-catenin/Tcf signaling induces the transcription of Axin2, a negative regulator of the signaling pathway. Molecular and Cellular Biology. 22 (4), 1172-1183 (2002).
  21. de Roo, J. J. D., et al. Axin2-mTurquoise2: A novel reporter mouse model for the detection of canonical Wnt signalling. Genesis. 55 (10), (2017).
  22. van de Moosdijk, A. A. A., van de Grift, Y. B. C., de Man, S. M. A., Zeeman, A. L., van Amerongen, R. A novel Axin2 knock-in mouse model for visualization and lineage tracing of WNT/CTNNB1 responsive cells. Genesis. 58 (9), 23387 (2020).
  23. Choi, Y. S., et al. Distinct functions for Wnt/beta-catenin in hair follicle stem cell proliferation and survival and interfollicular epidermal homeostasis. Cell Stem Cell. 13 (6), 720-733 (2013).
  24. Nolan, G. P., Fiering, S., Nicolas, J. F., Herzenberg, L. A. Fluorescence-activated cell analysis and sorting of viable mammalian cells based on beta-D-galactosidase activity after transduction of Escherichia coli lacZ. Proceedings of the National Academy of Sciences of the United States of America. 85 (8), 2603-2607 (1988).
  25. Goedhart, J., et al. Structure-guided evolution of cyan fluorescent proteins towards a quantum yield of 93. Nature Communications. 3, 751 (2012).
  26. Bernkopf, D. B., Bruckner, M., Hadjihannas, M. V., Behrens, J. An aggregon in conductin/axin2 regulates Wnt/beta-catenin signaling and holds potential for cancer therapy. Nat Commun. 10 (1), 4251 (2019).
  27. Joosen, L., Hink, M. A., Gadella, T. W., Goedhart, J. Effect of fixation procedures on the fluorescence lifetimes of Aequorea victoria derived fluorescent proteins. Journal of Microscopy. 256 (3), 166-176 (2014).
  28. Schindelin, J., et al. Fiji: an open-source platform for biological-image analysis. Nature Methods. 9 (7), 676-682 (2012).
  29. Monici, M. Cell and tissue autofluorescence research and diagnostic applications. Biotechnology Annual Reviews. 11, 227-256 (2005).
  30. Henriksson, J., et al. Endrov: an integrated platform for image analysis. Nature Methods. 10 (6), 454-456 (2013).
  31. Hedgepeth, C. M., et al. Activation of the Wnt signaling pathway: a molecular mechanism for lithium action. Developmental Biology. 185 (1), 82-91 (1997).
  32. Sato, N., Meijer, L., Skaltsounis, L., Greengard, P., Brivanlou, A. H. Maintenance of pluripotency in human and mouse embryonic stem cells through activation of Wnt signaling by a pharmacological GSK-3-specific inhibitor. Nature Medicine. 10 (1), 55-63 (2004).
  33. Ring, D. B., et al. Selective glycogen synthase kinase 3 inhibitors potentiate insulin activation of glucose transport and utilization in vitro and in vivo. Diabetes. 52 (3), 588-595 (2003).
  34. Liu, C., et al. Control of beta-catenin phosphorylation/degradation by a dual-kinase mechanism. Cell. 108 (6), 837-847 (2002).
  35. Zeng, X., et al. Initiation of Wnt signaling: control of Wnt coreceptor Lrp6 phosphorylation/activation via frizzled, dishevelled and axin functions. Development. 135 (2), 367-375 (2008).
This article has been published
Video Coming Soon
Keep me updated:

.

Cite This Article
de Roo, J. J. D., Naber, B. A., Vloemans, S. A., de Haas, E. F., van der Laan, A. M., Staal, F. J. Flow Cytometry and Confocal Imaging Analysis of Low Wnt Expression in Axin2-mTurquoise2 Reporter Thymocytes. J. Vis. Exp. (175), e62141, doi:10.3791/62141 (2021).

View Video