Summary

A Label-free Xenograft Model for Investigating the Behavior of Human Stem Cell Spheroids in Chick Embryos

Published: October 15, 2021
doi:

Summary

Here, we describe a method to transplant and identify human cell spheroids into chick embryos. This xenograft model uses the embryonic microenvironment as a source of instructive signals to assay cell migration, differentiation, and tropism and is especially suited for the study of primary and/or heterogeneous cell populations.

Abstract

Xenografts are valuable methods to investigate the behavior of human cells in vivo. In particular, the embryonic environment provides cues for cell migration, differentiation, and morphogenesis, with unique instructive signals and germ layer identity that are often absent from adult xenograft models. In addition, embryonic models cannot discriminate self versus non-self tissues, eliminating the risk of rejection of the graft and the need for immune suppression of the host. This paper presents a methodology for transplantation of spheroids of human cells into chicken embryos, which are accessible, amenable to manipulation, and develop at 37 °C.

Spheroids allow the selection of a specific region of the embryo for transplantation. After being grafted, the cells become integrated into the host tissue, allowing the follow-up of their migration, growth, and differentiation. This model is flexible enough to allow the utilization of different adherent populations, including heterogeneous primary cell populations and cancer cells. To circumvent the need for prior cell labeling, a protocol for the identification of donor cells through hybridization of human-specific Alu probes is also described, which is particularly important when investigating heterogeneous cell populations. Furthermore, DNA probes can be easily adapted to identify other donor species. This protocol will describe the general methods for preparing spheroids, grafting into chicken embryos, fixing and processing tissue for paraffin sectioning, and finally identifying the human cells using DNA in situ hybridization. Suggested controls, examples of interpretation of results and various cell behaviors that can be assayed will be discussed in addition to the limitations of this method.

Introduction

Xenografts are useful tools to investigate the behavior of human cells in vivo. These models have provided invaluable information for a wide range of scientific topics, such as the biology of human stem cells1, the observation of cellular events in real time2, and the investigation of tumoral angiogenesis and metastasis3. In addition, several aspects of cancer biology, including the tumorigenesis of patient-specific xenografts, have been studied4,5. Each of these xenograft models has their advantages and disadvantages and, thus, each one is better suited for specific scientific questions. Chick embryos are a popular developmental biology model as they are an accessible amniote model that is amenable to surgical manipulation. Heterologous grafts have allowed researchers to create precise fate maps6 or explore whether a trait is cell-autonomous or instructed by the environment7,8. A similar rationale allows the chick embryo to be used as a xenograft model to study the behavior of human cells.

The embryonic environment actively orchestrates tissue morphogenesis with migration and differentiation signals, as well as cell-cell interactions. Thus, compared to adult xenograft models, the embryo provides a more instructive milieu to assay the behavior of grafted cells, for example, by mimicking signals present in adult stem cell niches (e.g., BMPs, WNTs, NOTCH, and SHH9). In addition, the absence of an adaptive immune system during early development allows xenografts to be performed without the risk of an immune response or rejection of the donor tissue10. Previous studies have investigated xenografts of human cells into chicken embryos for this purpose. The neurogenic potential of human stem cells has been assayed after injection into the neural tube or blood vessels11 in addition to the integration of embryonic stem cells12 and induced pluripotent stem cells13 into the embryo. Human melanoma cells have also been studied using the chick's embryonic environment, which revealed links between their tumorigenesis and the behavior of neural crest cells14, as well as the reprogramming of the tumor cells with the information from the embryo15. This paper describes a protocol that is especially suited for studying the behavior of human primary and heterogeneous cell populations.

In the last decades, the stromal component of diverse tissues has been studied as an autologous source of progenitor/stem cells and for its proangiogenic and immunoregulatory properties, previously known as "mesenchymal stem cells"16,17,18. The first of these cell populations to be characterized was the bone marrow stromal/stem cell population (BMSCs), which have osteo-, adipo- and, to a lesser extent, chondrogenic potential in vivo19,20. Adipose-derived stromal cells (ADSCs) are a heterogeneous population obtained by enzymatic digestion of the lipoaspirate or dermolipectomy samples, followed by isolation of the stromal-vascular fraction (SVF) and finally expansion in culture21. In culture, these cells are phenotypically characterized by markers shared with other mesenchymal populations, such as CD90, CD73, CD105, and CD44, unique markers such as CD36, and the absence of hematopoietic (CD45) or endothelial (CD31) markers22. Additionally, ADSCs have osteo-, adipo-, and chondrogenic potential in vitro, and the number of stem/progenitor cells in this population can be defined by the fibroblastoid colony-forming unit (CFU-F) assay22. In vivo, cells with the ADSC phenotype have been reported to exist in stromal23 and/or perivascular24 compartments. It is becoming increasingly clear that, despite sharing markers after in vitro culture, the stromal compartment of different tissues reflects intrinsic characteristics of a given organ, and these cell populations have distinct properties depending on their source17,25,26,27. Furthermore, as these cells are isolated based on their adhesion to a cell culture dish, they may be composed of cells from diverse germ layers28. Thus, employing a xenograft method to study the differentiation potential and tropism of stromal cells in an unbiased way can provide valuable information about these cell populations to guide the development of future cell therapies.

The protocol described here (Figure 1) is a xenograft method that takes advantage of the low cost and ease of manipulation of chick embryos. It has been previously used to study the behavior of human ADSC29, skin fibroblasts29, menstrual blood-derived stromal cells30, and glioblastoma cells31. This method will include the transplantation of cells as spheroids32, which can be prepared from any population of adherent cells (Figure 2). Surgical procedures and the preparation of custom surgical materials-the microscalpels and glass capillaries-will also be described (Figure 3). Human cells are detected in histological sections by hybridizing human-specific Alu probes (Figure 4), thus eliminating the need for prior labeling of the grafted cells. The representative results describe the behavior of human ADSC grafted both in the somitic region at the wing bud level (Figure 5, Figure 6, and Figure 7) and the first pharyngeal arch (Figure 8), as well as human primary glioblastoma spheroids grafted in the prosencephalon (Figure 8). Cell migration, differentiation, and interaction with chick embryonic tissues will be described, as well as suggested assays to further investigate cell behavior using co-staining or staining of adjacent sections.

Protocol

All in vivo procedures used in this study complied with all relevant experimental guidelines for animal testing and research, in accordance with the Brazilian experimental animal use guidelines (L11794). The protocols used for handling chicken embryos were all approved by the Ethics Committee on the Use of Animals in Scientific Experimentation (Health Sciences Centre of the Federal University of Rio de Janeiro). The use of human cells was approved by the Ethics Committee of the University Hospital Clementino Fra…

Representative Results

Identification of Alu-positive ADSCs in histological sections Alu sequences are repetitive elements that comprise ~10% of the human genome and thus are excellent targets for identifying human cells in a species-specific manner43. In situ hybridization with DNA probes can be used to identify genomic elements on histological sections, including primary human cells29,<sup class="xref…

Discussion

The protocol described here (Figure 1) presents a feasible option for screening the behavior of primary populations of human cells in vivo, using chick embryos as a model. This paper describes the formation of cell spheroids (Figure 2), transplantation of the spheroid into the chick embryo (Figure 3), processing of specimens and in situ hybridization (Figure 4), representative results …

Disclosures

The authors have nothing to disclose.

Acknowledgements

This work was supported by Universidade Federal de Rio de Janeiro (UFRJ for J.B.), Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq for J.B.) and Fundação Carlos Chagas Filho de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ for J.B.). We thank T. Jaffredo (CNRS, Paris, France) for the Runx2 (Cbfa1) probe. The HNK1 antibody was obtained from the Developmental Studies Hybridoma Bank developed under the auspices of the NICHD and maintained by The University of Iowa, Department of Biological Sciences, Iowa City, IA 52242 USA. We thank V. Moura-Neto for granting access to the microtome and R. Lent for granting access to the microscope. We thank E. Steck for the help in synthesizing Alu probes.

Materials

Animals
Gallus gallus eggs Granja Tolomei SPF-free White leghorn chicken
Reagents
Alcian Blue 8GX Sigma-aldrich A5268
AluFw primers Sigma-aldrich OLIGO 5’-CGA GGC GGG TGG ATC ATG AGG T-3’
AluRev primers Sigma-aldrich OLIGO 5’-TTT TTT GAG ACG GAG TCT CGC-3’
Aluminum sulphate Sigma-aldrich 368458 For Nuclear fast red solution preparation
Anti-Digoxigenin-AP, Fab fragments Roche 11093274910 Antibody Registry ID: AB_514497
Anti-Human Natural Killer 1 antibody (HNK1, CD57) Developmental Studies Hybridoma Bank 3H5 Antibody Registry ID: AB_2314644
Anti-mouse, goat IgM-HRP Santa Cruz Biotechnology sc-2973 Antibody Registry ID: AB_650513
Anti-mouse, goat IgG (H+L)-HRP Novex G-21040 Antibody Registry ID: AB_2536527
Anti-Smooth Muscle Actin/ACTA2 antibody Dako M085129 Antibody Registry ID: AB_2811108
Aquatex Merck 1085620050 Aqueous mounting agent
5-Bromo-4-chloro-3-indolyl phosphate p-toluidine salt (BCIP) Sigma-aldrich B8503-100MG
Blocking Reagent Roche 11096176001
Citric acid VETEC 238 For SSC buffer preparation
Collagenase type IA Sigma-aldrich SCR103
dCTP, dGTP, dATP, dTTP set Roche 11969064001
Denhardt solution 50X Invitrogen 750018 For hybridization buffer preparation
Dextran sulphate sodium salt Thermo Scientific 15885118 For hybridization buffer preparation
DIG RNA Labeling Mix Roche 11277073910 Contains Dig-11-dUTP
DMEM low-glucose Sigma-aldrich D5523
3,3′-Diaminobenzidine tetrahydrochloride (DAB) Sigma-aldrich D5905-50TAB
N,N-Dimethylformamide (DMF) Sigma-aldrich 227056 For NBT and BCIP solution preparation
Ethylenediaminetetraacetic acid (EDTA) Sigma-aldrich E6758 For trypsin solution preparation
Entellan new Merck 107961 Non-aqueous mounting medium
Ethanol Proquímios N/A
Fetal bovine serum ThermoFisher 12657029 Inactivate at 56 °C before use
Formaldehyde 37% solution Proquímios N/A
Formamide Vetec V900064
Glacial acetic acid Proquímios N/A
India ink Pelikan 221143
L-glutamine solution (200 mM) Gibco 25030-149
Magnesium chloride Merck 8147330100 For NTM buffer preparation
Maleic acid Sigma-aldrich M0375-500G For MAB buffer preparation
Methanol Proquímios
Normal Goat Serum Sigma-aldrich NS02L Inactivate at 56 °C before use
4-Nitro blue tetrazolium chloride (NBT) Roche 11585029001
Nuclear fast red Sigma-aldrich 60700
Paraplast Plus Sigma-aldrich P3558
Penicillin G sodium salt Sigma-aldrich P3032
Phosphate buffered saline (PBS) Sigma-aldrich P3813
Phosphomolybdic acid Merck 100532
Proteinase K Gibco BRL 25530-015
Salmon sperm DNA Invitrogen 15632011 For hybridization buffer preparation
Sodium chloride Sigma-aldrich S9888 For SSC, MAB and NTM buffer preparation
Streptomycin Sulfate Sigma-aldrich S6501
Taq Polymerase kit Cenbiot Enzimas N/A
Tris-HCl Sigma-aldrich T5941
Trypsin Sigma-aldrich T4799
Tween 20 Sigma-aldrich P1379
Xylene Proquímios N/A
Microscope and equipments
Axioplan upright microscope Carl Zeiss Microscopy N/A
Axiovision software Carl Zeiss Microscopy N/A
Cell incubator ThermoForma 3110
Egg incubator- 50 eggs GP
Gooseneck lamp Biocam N/A For egg manipulation
Fiji software; Cell Counter plugin ImageJ https://imagej.net/software/fiji/
Laminar flow hood TROX 1385
Nanodrop Lite Thermo Scientific ND-LITE-PR
Rotary microtome Leica Biosystems RM2125 RTS For sectioning
Stereomicroscope Labomed Luxeo 4D For egg manipulation
Sterilization oven REALIS 7261690 For sterelization of surgical materials
Consumables
0.2 mL (PCR) polypropylene centrifuge tubes Eppendorf 30124707
15 mL polypropylene conical centrifuge tubes Corning CLS430791
1.5 mL polypropylene centrifuge tubes Axygen MCT-150-C
2 mL polypropylene centrifuge tubes Axygen MCT-200-C
50 mL polypropylene conical centrifuge tubes Corning CLS430829
Barrier (Filter) Tips, 200 μL size Invitrogen AM12655 For egg manipulation
Excavated Glass Block (Staining Block) with Cover Glass Hecht Karl 42020010
Embedding cassettes Simport M480 Used as a paraffin block holder
Glass coverslides, 24 x 40 mm Kasvi K5-2440
Glass Pasteur pipettes 230 mm NORMAX 5426023 For preparation of glass capillaries
Microtome blades Leica Biosystems HIGH-PROFILE-DISPOSABLE-BLADES-818 For sectioning
Parafilm M Parafilm P7793
Plastic Petri dish, 30 mm Kasvi K13-0035 For egg manipulation
Plastic Petri dish, 60 mm Prolab 0303-8 For cell spheroids preparation. Should not be treated for cell adhesion. 
Silanized glass slides (Starfrost) Knittel Glass 198 For sectioning
Syringe 1 mL , Needles 26 G (0.45 x 13 mm) Descarpack 32972 For egg manipulation (albumen aspiration)
Surgical tools
Aspirator tube Drummond 2-000-000 For egg manipulation
Dissection scissors Fine Science Tools 14061-11 For egg manipulation
Microforceps (tweezers) Fine Science Tools 00108-11 For egg manipulation and preparation of glass capillaries
Needle holder (adjustable dissection needle chuck) Fisherbrand 8955 For egg manipulation
Oil whetstone, 10.000 grit N/A N/A For sharpening needles
Pair of small paint brushes N/A N/A For handling paraffin sections. Any brand may be used.
Sewing needles N/A N/A For sharpening into microscalpels. Any brand may be used.
Sterile disposable scalpel No. 23 Swann-Norton 110 For sectioning
Surgical scalpel handle Swann-Norton 914 For sectioning
Wecker iris scissors, sharp/sharp Surtex SS-641-11 For egg manipulation

References

  1. Herbert, K. E., Lévesque, J. P., Haylock, D. N., Prince, M. The use of experimental murine models to assess novel agents of hematopoietic stem and progenitor cell mobilization. Biology of Blood and Marrow Transplantation. 14 (6), 603-621 (2008).
  2. Parada-Kusz, M., et al. Generation of mouse-zebrafish hematopoietic tissue chimeric embryos for hematopoiesis and host-pathogen interaction studies. DMM Disease Models and Mechanisms. 11 (11), (2018).
  3. Lokman, N. A., Elder, A. S. F., Ricciardelli, C., Oehler, M. K. Chick chorioallantoic membrane (CAM) assay as an in vivo model to study the effect of newly identified molecules on ovarian cancer invasion and metastasis. International Journal of Molecular Sciences. 13 (8), 9959-9970 (2012).
  4. Jung, J. Human tumor xenograft models for preclinical assessment of anticancer drug development. Toxicological Research. 30 (1), 1-5 (2014).
  5. Ben-David, U., et al. Patient-derived xenografts undergo mouse-specific tumor evolution. Nature Genetics. 49 (11), 1567-1575 (2017).
  6. le Douarin, N., Kalcheim, C. . The Neural Crest. , (1999).
  7. Schneider, R. A. Neural crest and the origin of species-specific pattern. Genesis. 56 (6-7), 23219 (2018).
  8. Santagati, F., Rijli, F. M. Cranial neural crest and the building of the vertebrate head. Nature Reviews Neuroscience. 4 (10), 806-818 (2003).
  9. Li, L., Clevers, H. Coexistence of quiescent and active adult stem cells in mammals. Science. 327 (5965), 542-545 (2010).
  10. Douarin, N., et al. le et al. Evidence for a thymus-dependent form of tolerance that is not based on elimination or anergy of reactive T cells. Immunological Reviews. 149 (1), 35-53 (1996).
  11. Boulland, J. L., Halasi, G., Kasumacic, N., Glover, J. C. Xenotransplantation of human stem cells into the chicken embryo. Journal of Visualized Experiments: JoVE. (41), e2071 (2010).
  12. Goldstein, R. S. Transplantation of human embryonic stem cells and derivatives to the chick embryo. Methods in Molecular Biology. 584, 367-385 (2010).
  13. Akhlaghpour, A., et al. Chicken interspecies chimerism unveils human pluripotency. Stem Cell Reports. 16 (1), 39-55 (2021).
  14. Kulesa, P. M., Morrison, J. A., Bailey, C. M. The neural crest and cancer: A developmental spin on melanoma. Cells Tissues Organs. 198 (1), 12-21 (2013).
  15. Hendrix, M. J. C., et al. Reprogramming metastatic tumour cells with embryonic microenvironments. Nature Reviews Cancer. 7 (4), 246-255 (2007).
  16. Singer, N. G., Caplan, A. I. Mesenchymal stem cells: Mechanisms of inflammation. Annual Review of Pathology: Mechanisms of Disease. 6, 457-478 (2011).
  17. Bianco, P., Robey, P. G., Simmons, P. J. Mesenchymal stem cells: Revisiting history, concepts, and assays. Cell Stem Cell. 2 (4), 313-319 (2008).
  18. Griffin, M. D., et al. Concise review: Adult mesenchymal stromal cell therapy for inflammatory diseases: How well are we joining the dots. Stem Cells. 31 (10), 2033-2041 (2013).
  19. Owen, M., Friedenstein, A. J. Stromal stem cells: marrow-derived osteogenic precursors. Ciba Foundation Symposium. 136, 42-60 (1988).
  20. Bianco, P., Robey, P. G., Saggio, I., Riminucci, M. “Mesenchymal” stem cells in human bone marrow (skeletal stem cells): A critical discussion of their nature, identity, and significance in incurable skeletal disease. Human Gene Therapy. 21 (9), 1057-1066 (2010).
  21. Zuk, P. A., et al. Multilineage cells from human adipose tissue: Implications for cell-based therapies. Tissue Engineering. 7 (2), 211-228 (2001).
  22. Bourin, P., et al. Stromal cells from the adipose tissue-derived stromal vascular fraction and culture expanded adipose tissue-derived stromal/stem cells: A joint statement of the International Federation for Adipose Therapeutics and Science (IFATS) and the International Society for Cellular Therapy (ISCT). Cytotherapy. 15 (6), 641-648 (2013).
  23. Maumus, M., et al. Native human adipose stromal cells: Localization, morphology and phenotype. International Journal of Obesity. 35 (9), 1141-1153 (2011).
  24. Zannettino, A. C. W., et al. Multipotential human adipose-derived stromal stem cells exhibit a perivascular phenotype in vitro and in vivo. Journal of Cellular Physiology. 214 (2), 413-421 (2008).
  25. Phinney, D. G. Functional heterogeneity of mesenchymal stem cells: Implications for cell therapy. Journal of Cellular Biochemistry. 113 (9), 2806-2812 (2012).
  26. Dominici, M., et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 8 (4), 315-317 (2006).
  27. Baptista, L. S., et al. marrow and adipose tissue-derived mesenchymal stem cells: How close are they. Journal of Stem Cells. , 73-90 (2007).
  28. Sowa, Y., et al. Adipose stromal cells contain phenotypically distinct adipogenic progenitors derived from neural crest. PLoS ONE. 8 (12), 84206 (2013).
  29. Cordeiro, I. R., et al. Chick embryo xenograft model reveals a novel perineural niche for human adipose-derived stromal cells. Biology Open. 4 (9), 1180-1193 (2015).
  30. Santos, R. d. e. A., et al. Intrinsic angiogenic potential and migration capacity of human mesenchymal stromal cells derived from menstrual blood and bone marrow. International Journal of Molecular Sciences. 21 (24), 1-23 (2020).
  31. Menezes, A., et al. Live cell imaging supports a key role for histone deacetylase as a molecular target during glioblastoma malignancy downgrade through tumor competence modulation. Journal of Oncology. 2019, 9043675 (2019).
  32. Brito, J. M., Teillet, M. A., le Douarin, N. M. Induction of mirror-image supernumerary jaws in chicken mandibular mesenchyme by Sonic Hedgehog-producing cells. Development. 135 (13), 2311-2319 (2008).
  33. Foty, R. A simple hanging drop cell culture protocol for generation of 3D spheroids. Journal of Visualized Experiments JoVE. (51), e2720 (2011).
  34. de Barros, A. P. D. N., et al. Osteoblasts and bone marrow mesenchymal stromal cells control hematopoietic stem cell migration and proliferation in 3D in vitro model. PLoS One. 5 (2), 9093 (2010).
  35. Hamburger, V., Hamilton, H. L. A series of normal stages in the development of the chick embryo. Developmental Dynamics. 195 (4), 231-272 (1951).
  36. Brady, J. A simple technique for making very fine, durable dissecting needles by sharpening tungsten wire electrolytically. Bulletin of the World Health Organization. 32 (1), 143-144 (1965).
  37. Tickle, C. How the embryo makes a limb: Determination, polarity and identity. Journal of Anatomy. 227 (4), 418-430 (2015).
  38. Couly, G., Grapin-Botton, A., Coltey, P., le Douarin, N. M. The regeneration of the cephalic neural crest, a problem revisited: The regenerating cells originate from the contralateral or from the anterior and posterior neural fold. Development. 122 (11), 3393-3407 (1996).
  39. Walker, J. A., et al. Human DNA quantitation using Alu element-based polymerase chain reaction. Analytical Biochemistry. 315 (1), 122-128 (2003).
  40. Steck, E., Burkhardt, M., Ehrlich, H., Richter, W. Discrimination between cells of murine and human origin in xenotransplants by species specific genomic in situ hybridization. Xenotransplantation. 17 (2), 153-159 (2010).
  41. Sams, A., Davies, F. M. R. Commercial varieties of nuclear fast red. Stain Technology. 42 (6), 269-276 (1967).
  42. Lison, L. Alcian blue 8 g with chlorantine fast red 5 B. A technic for selective staining of mucopolysaccharides. Biotechnic and Histochemistry. 29 (3), 131-138 (1954).
  43. Cordaux, R., Batzer, M. A. The impact of retrotransposons on human genome evolution. Nature Reviews Genetics. 10 (10), 691-703 (2009).
  44. Brüstle, O., et al. Chimeric brains generated by intraventricular transplantation of fetal human brain cells into embryonic rats. Nature Biotechnology. 16 (11), 1040-1044 (1998).
  45. Warncke, B., Valtink, M., Weichel, J., Engelmann, K., Schäfer, H. Experimental rat model for therapeutic retinal pigment epithelium transplantation – Unequivocal microscopic identification of human donor cells by in situ hybridisation of human-specific Alu sequences. Virchows Archiv. 444 (1), 74-81 (2004).
  46. Kasten, P., et al. Ectopic bone formation associated with mesenchymal stem cells in a resorbable calcium deficient hydroxyapatite carrier. Biomaterials. 26 (29), 5879-5889 (2005).
  47. Baptista, L., et al. Adipose tissue of control and ex-obese patients exhibit differences in blood vessel content and resident mesenchymal stem cell population. Obesity Surgery. 19 (9), 1304-1312 (2009).
  48. Christ, B., Huang, R., Scaal, M. Formation and differentiation of the avian sclerotome. Anatomy and Embryology. 208 (5), 333-350 (2004).
  49. Scaal, M., Christ, B. Formation and differentiation of the avian dermomyotome. Anatomy and Embryology. 208 (6), 411-424 (2004).
  50. Tucker, G. C., Delarue, M., Zada, S., Boucaut, J. C., Thiery, J. P. Expression of the HNK-1/NC-1 epitope in early vertebrate neurogenesis. Cell and Tissue Research. 251 (2), 457-465 (1988).
  51. Creuzet, S., Schuler, B., Couly, G., le Douarin, N. M. Reciprocal relationships between Fgf8 and neural crest cells in facial and forebrain development. Proceedings of the National Academy of Sciences of the United States of America. 101 (14), 4843-4847 (2004).
  52. Charrier, J. B., Lapointe, F., le Douarin, N. M., Teillet, M. A. Dual origin of the floor plate in the avian embryo. Development. 129 (20), 4785-4796 (2002).
  53. Kordes, U., Cheng, Y. C., Scotting, P. J. Sox group e gene expression distinguishes different types and maturational stages of glial cells in developing chick and mouse. Developmental Brain Research. 157 (2), 209-213 (2005).
  54. Stricker, S., Fundele, R., Vortkamp, A., Mundlos, S. Role of Runx genes in chondrocyte differentiation. Developmental Biology. 245 (1), 95-108 (2002).
  55. Grottkau, B. E., Lin, Y. Osteogenesis of adipose-derived stem cells. Bone Research. 1 (2), 133 (2013).
  56. Culling, C. F. A. . Handbook of histopathological and histochemical techniques. Including museum techniques. , (1974).
  57. Francis, P. H., Richardson, M. K., Brickell, P. M., Tickle, C. Bone morphogenetic proteins and a signalling pathway that controls patterning in the developing chick limb. Development. 120 (1), 209-218 (1994).
  58. Huber, K., et al. Persistent expression of BMP-4 in embryonic chick adrenal cortical cells and its role in chromaffin cell development. Neural Development. 3, 28 (2008).
  59. Pouget, C., Gautier, R., Teillet, M. A., Jaffredo, T. Somite-derived cells replace ventral aortic hemangioblasts and provide aortic smooth muscle cells of the trunk. Development. 133 (6), 1013-1022 (2006).
  60. Kirby, M. L., Hutson, M. R. Factors controlling cardiac neural crest cell migration. Cell Adhesion and Migration. 4 (4), 609-621 (2010).
  61. Isern, J., et al. The neural crest is a source of mesenchymal stem cells with specialized hematopoietic stem cell niche function. eLife. 3, 03696 (2014).
  62. Yamazaki, S., et al. Nonmyelinating schwann cells maintain hematopoietic stem cell hibernation in the bone marrow niche. Cell. 147 (5), 1146-1158 (2011).
  63. Carr, M. J., et al. Mesenchymal precursor cells in adult nerves contribute to mammalian tissue repair and regeneration. Cell Stem Cell. 24 (2), 240-256 (2019).
  64. Walker, J. A., et al. Quantitative intra-short interspersed element PCR for species-specific DNA identification. Analytical Biochemistry. 316 (2), 259-269 (2003).
  65. Shultz, L. D., Ishikawa, F., Greiner, D. L. Humanized mice in translational biomedical research. Nature Reviews Immunology. 7 (2), 118-130 (2007).
  66. Yong, K. S. M., Her, Z., Chen, Q. Humanized mice as unique tools for human-specific studies. Archivum Immunologiae et Therapiae Experimentalis. 66 (4), 245-266 (2018).
  67. Cordeiro, I. R., Tanaka, M. Environmental oxygen is a key modulator of development and evolution: From molecules to ecology: Oxygen-sensitive pathways pattern the developing organism, linking genetic and environmental components during the evolution of new traits. BioEssays. 42 (9), 2000025 (2020).
  68. Poss, K. D., Wilson, L. G., Keating, M. T. Heart regeneration in zebrafish. Science. 298 (5601), 2188-2190 (2002).
  69. Puente, B. N., et al. The oxygen-rich postnatal environment induces cardiomyocyte cell-cycle arrest through DNA damage response. Cell. 157 (3), 565-579 (2014).
  70. Colton, C. K. Implantable biohybrid artificial organs. Cell Transplantation. 4 (4), 415-436 (1995).
  71. Meehan, G. R., et al. Developing a xenograft model of human vasculature in the mouse ear pinna. Scientific Reports. 10 (1), 2058 (2020).
This article has been published
Video Coming Soon
Keep me updated:

.

Cite This Article
Rosenburg Cordeiro, I., de Brito Neto, J. M. A Label-free Xenograft Model for Investigating the Behavior of Human Stem Cell Spheroids in Chick Embryos. J. Vis. Exp. (176), e63067, doi:10.3791/63067 (2021).

View Video