Skip to main content
Log in

In Vitro and In Vivo Cardiomyogenic Differentiation of Amniotic Fluid Stem Cells

  • Published:
Stem Cell Reviews and Reports Aims and scope Submit manuscript

Abstract

Cell therapy has developed as a complementary treatment for myocardial regeneration. While both autologous and allogeneic uses have been advocated, the ideal candidate has not been identified yet. Amniotic fluid-derived stem (AFS) cells are potentially a promising resource for cell therapy and tissue engineering of myocardial injuries. However, no information is available regarding their use in an allogeneic context. c-kit-sorted, GFP-positive rat AFS (GFP-rAFS) cells and neonatal rat cardiomyocytes (rCMs) were characterized by cytocentrifugation and flow cytometry for the expression of mesenchymal, embryonic and cell lineage-specific antigens. The activation of the myocardial gene program in GFP-rAFS cells was induced by co-culture with rCMs. The stem cell differentiation was evaluated using immunofluorescence, RT-PCR and single cell electrophysiology. The in vivo potential of Endorem-labeled GFP-rAFS cells for myocardial repair was studied by transplantation in the heart of animals with ischemia/reperfusion injury (I/R), monitored by magnetic resonance imaging (MRI). Three weeks after injection a small number of GFP-rAFS cells acquired an endothelial or smooth muscle phenotype and to a lesser extent CMs. Despite the low GFP-rAFS cells count in the heart, there was still an improvement of ejection fraction as measured by MRI. rAFS cells have the in vitro propensity to acquire a cardiomyogenic phenotype and to preserve cardiac function, even if their potential may be limited by poor survival in an allogeneic setting.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7

References

  1. Gonzales, C., & Pedrazzini, T. (2009). Progenitor cell therapy for heart disease. Experimental Cell Research, 315(18), 3077–3085.

    Article  CAS  PubMed  Google Scholar 

  2. Menasche, P. (2009). Cell-based therapy for heart disease: a clinically oriented perspective. Molecular Therapy, 17(5), 758–766.

    Article  CAS  PubMed  Google Scholar 

  3. Shintani, Y., Fukushima, S., Varela-Carver, A., et al. (2009). Donor cell-type specific paracrine effects of cell transplantation for post-infarction heart failure. Journal of Molecular and Cellular Cardiology, 47(2), 288–295.

    Article  CAS  PubMed  Google Scholar 

  4. Reinecke, H., Minami, E., Zhu, W. Z., & Laflamme, M. A. (2008). Cardiogenic differentiation and transdifferentiation of progenitor cells. Circulation Research, 103(10), 1058–1071.

    Article  CAS  PubMed  Google Scholar 

  5. Ausoni, S., & Sartore, S. (2009). From fish to amphibians to mammals: in search of novel strategies to optimize cardiac regeneration. The Journal of Cell Biology, 184(3), 357–364.

    Article  CAS  PubMed  Google Scholar 

  6. Oh, H., Chi, X., Bradfute, S. B., et al. (2004). Cardiac muscle plasticity in adult and embryo by heart-derived progenitor cells. Annals of the New York Academy of Sciences, 1015, 182–189.

    Article  PubMed  Google Scholar 

  7. Mummery, C., Ward-van, O. D., Doevendans, P., et al. (2003). Differentiation of human embryonic stem cells to cardiomyocytes: role of coculture with visceral endoderm-like cells. Circulation, 107(21), 2733–2740.

    Article  CAS  PubMed  Google Scholar 

  8. DeCoppi, P., Bartsch, G., Jr., Siddiqui, M. M., et al. (2007). Isolation of amniotic stem cell lines with potential for therapy. Nature Biotechnology, 25(1), 100–106.

    Article  CAS  Google Scholar 

  9. DeCoppi, P., Callegari, A., Chiavegato, A., et al. (2007). Amniotic fluid and bone marrow derived mesenchymal stem cells can be converted to smooth muscle cells in the cryo-injured rat bladder and prevent compensatory hypertrophy of surviving smooth muscle cells. Journal d'Urologie, 177(1), 369–376.

    Article  Google Scholar 

  10. Mauro, A., Turriani, M., Ioannoni, A., et al. (2010). Isolation, characterization, and in vitro differentiation of ovine amniotic stem cells. Veterinary Research Communications, 34(Suppl 1), S25–S28.

    Article  PubMed  Google Scholar 

  11. Gekas, J., Walther, G., Skuk, D., Bujold, E., Harvey, I., & Bertrand, O. F. (2010). In vitro and in vivo study of human amniotic fluid-derived stem cell differentiation into myogenic lineage. Clinical and Experimental Medicine, 10(1), 1–6.

    Article  PubMed  Google Scholar 

  12. Fujimoto, K. L., Miki, T., Liu, L. J., et al. (2009). Naive rat amnion-derived cell transplantation improved left ventricular function and reduced myocardial scar of postinfarcted heart. Cell Transplantation, 18(4), 477–486.

    Article  PubMed  Google Scholar 

  13. Zhao, P., Ise, H., Hongo, M., Ota, M., Konishi, I., & Nikaido, T. (2005). Human amniotic mesenchymal cells have some characteristics of cardiomyocytes. Transplantation, 79(5), 528–535.

    Article  PubMed  Google Scholar 

  14. Okamoto, K., Miyoshi, S., Toyoda, M., et al. (2007). ‘Working’ cardiomyocytes exhibiting plateau action potentials from human placenta-derived extraembryonic mesodermal cells. Experimental Cell Research, 313(12), 2550–2562.

    Article  CAS  PubMed  Google Scholar 

  15. Sartore, S., Lenzi, M., Angelini, A., et al. (2005). Amniotic mesenchymal cells autotransplanted in a porcine model of cardiac ischemia do not differentiate to cardiogenic phenotypes. European Journal of Cardiothoracic Surgery, 28(5), 677–684.

    Article  PubMed  Google Scholar 

  16. Chiavegato, A., Bollini, S., Pozzobon, M., et al. (2007). Human amniotic fluid-derived stem cells are rejected after transplantation in the myocardium of normal, ischemic, immuno-suppressed or immuno-deficient rat. Journal of Molecular and Cellular Cardiology, 42(4), 746–759.

    Article  CAS  PubMed  Google Scholar 

  17. Iop, L., Chiavegato, A., Callegari, A., et al. (2008). Different cardiovascular potential of adult- and fetal-type mesenchymal stem cells in a rat model of heart cryoinjury. Cell Transplantation, 17(6), 679–694.

    Article  PubMed  Google Scholar 

  18. Guan, X., Delo, D. M., Atala, A., & Soker, S. (2010). In vitro cardiomyogenic potential of human amniotic fluid stem cells. J Tissue Eng Regen Med.

  19. Yeh, Y. C., Lee, W. Y., Yu, C. L., et al. (2010). Cardiac repair with injectable cell sheet fragments of human amniotic fluid stem cells in an immune-suppressed rat model. Biomaterials, 31(25), 6444–6453.

    Article  CAS  PubMed  Google Scholar 

  20. Li, C. D., Zhang, W. Y., Li, H. L., et al. (2005). Mesenchymal stem cells derived from human placenta suppress allogeneic umbilical cord blood lymphocyte proliferation. Cell Research, 15(7), 539–547.

    Article  CAS  PubMed  Google Scholar 

  21. Chang, C. J., Yen, M. L., Chen, Y. C., et al. (2006). Placenta-derived multipotent cells exhibit immunosuppressive properties that are enhanced in the presence of interferon-gamma. Stem Cells, 24(11), 2466–2477.

    Article  CAS  PubMed  Google Scholar 

  22. Magatti, M., De, M. S., Vertua, E., Gibelli, L., Wengler, G. S., & Parolini, O. (2008). Human amnion mesenchyme harbors cells with allogeneic T-cell suppression and stimulation capabilities. Stem Cells, 26(1), 182–192.

    Article  CAS  PubMed  Google Scholar 

  23. Banas, R. A., Trumpower, C., Bentlejewski, C., Marshall, V., Sing, G., & Zeevi, A. (2008). Immunogenicity and immunomodulatory effects of amnion-derived multipotent progenitor cells. Human Immunology, 69(6), 321–328.

    Article  CAS  PubMed  Google Scholar 

  24. Ditadi, A., DeCoppi, P., Picone, O., et al. (2009). Human and murine amniotic fluid c-Kit + Lin- cells display hematopoietic activity. Blood, 113(17), 3953–3960.

    Article  CAS  PubMed  Google Scholar 

  25. Dobreva, M. P., Pereira, P. N., Deprest, J., & Zwijsen, A. (2010). On the origin of amniotic stem cells: of mice and men. The International Journal of Developmental Biology, 54(5), 761–777.

    Article  CAS  PubMed  Google Scholar 

  26. Radisic, M., Park, H., Shing, H., et al. (2004). Functional assembly of engineered myocardium by electrical stimulation of cardiac myocytes cultured on scaffolds. Proceedings of the National Academy of Sciences of the United States of America, 101(52), 18129–18134.

    Article  CAS  PubMed  Google Scholar 

  27. Riegler, J., Wells, J. A., Kyrtatos, P. G., Price, A. N., Pankhurst, Q. A., & Lythgoe, M. F. (2010). Targeted magnetic delivery and tracking of cells using a magnetic resonance imaging system. Biomaterials, 31(20), 5366–5371.

    Article  CAS  PubMed  Google Scholar 

  28. Heiberg, E., Sjogren, J., Ugander, M., Carlsson, M., Engblom, H., & Arheden, H. (2010). Design and validation of Segment–freely available software for cardiovascular image analysis. BMC Medical Imaging, 10, 1.

    Article  PubMed  Google Scholar 

  29. Beltrami, A. P., Barlucchi, L., Torella, D., et al. (2003). Adult cardiac stem cells are multipotent and support myocardial regeneration. Cell, 114(6), 763–776.

    Article  CAS  PubMed  Google Scholar 

  30. Miyamoto, S., Kawaguchi, N., Ellison, G. M., Matsuoka, R., Shin’oka, T., & Kurosawa, H. (2010). Characterization of long-term cultured c-kit + cardiac stem cells derived from adult rat hearts. Stem Cells and Development, 19(1), 105–116.

    Article  CAS  PubMed  Google Scholar 

  31. Rogers, W. J., Meyer, C. H., & Kramer, C. M. (2006). Technology insight: in vivo cell tracking by use of MRI. Nature Clinical Practice. Cardiovascular Medicine, 3(10), 554–562.

    Article  CAS  PubMed  Google Scholar 

  32. Weisskoff, R. M., Zuo, C. S., Boxerman, J. L., & Rosen, B. R. (1994). Microscopic susceptibility variation and transverse relaxation: theory and experiment. Magnetic Resonance in Medicine, 31(6), 601–610.

    Article  CAS  PubMed  Google Scholar 

  33. Stuckey, D. J., Carr, C. A., Martin-Rendon, E., et al. (2006). Iron particles for noninvasive monitoring of bone marrow stromal cell engraftment into, and isolation of viable engrafted donor cells from, the heart. Stem Cells, 24(8), 1968–1975.

    Article  CAS  PubMed  Google Scholar 

  34. Kadner, A., Hoerstrup, S. P., Tracy, J., et al. (2002). Human umbilical cord cells: a new cell source for cardiovascular tissue engineering. The Annals of Thoracic Surgery, 74(4), S1422–S1428.

    Article  PubMed  Google Scholar 

  35. Schmidt, D., Breymann, C., Weber, A., et al. (2004). Umbilical cord blood derived endothelial progenitor cells for tissue engineering of vascular grafts. The Annals of Thoracic Surgery, 78(6), 2094–2098.

    Article  PubMed  Google Scholar 

  36. Yen, B. L., Huang, H. I., Chien, C. C., et al. (2005). Isolation of multipotent cells from human term placenta. Stem Cells, 23(1), 3–9.

    Article  CAS  PubMed  Google Scholar 

  37. Miao, Z., Jin, J., Chen, L., et al. (2006). Isolation of mesenchymal stem cells from human placenta: comparison with human bone marrow mesenchymal stem cells. Cell Biology International, 30(9), 681–687.

    Article  CAS  PubMed  Google Scholar 

  38. Chan, J., Kennea, N. L., & Fisk, N. M. (2007). Placental mesenchymal stem cells. American Journal of Obstetrics and Gynecology, 196(2), e18–e19.

    Article  PubMed  Google Scholar 

  39. Wang, M., Yang, Y., Yang, D., et al. (2009). The immunomodulatory activity of human umbilical cord blood-derived mesenchymal stem cells in vitro. Immunology, 126(2), 220–232.

    Article  CAS  PubMed  Google Scholar 

  40. Kadner, A., Hoerstrup, S. P., Tracy, J., et al. (2002). Human umbilical cord cells: a new cell source for cardiovascular tissue engineering. The Annals of Thoracic Surgery, 74(4), S1422–S1428.

    Article  PubMed  Google Scholar 

  41. Prusa, A. R., Marton, E., Rosner, M., Bernaschek, G., & Hengstschlager, M. (2003). Oct-4-expressing cells in human amniotic fluid: a new source for stem cell research? Human Reproduction, 18(7), 1489–1493.

    Article  PubMed  Google Scholar 

  42. Perin, L., Sedrakyan, S., DaSacco, S., & DeFilippo, R. (2008). Characterization of human amniotic fluid stem cells and their pluripotential capability. Methods in Cell Biology, 86, 85–99.

    Article  CAS  PubMed  Google Scholar 

  43. Perin, L., Giuliani, S., Sedrakyan, S., DaSacco, S., & DeFilippo, R. E. (2008). Stem Cell and Regenerative Science Applications in the Development of Bioengineering of Renal Tissue. Pediatr Res.

  44. Simantov, R. (2008). Amniotic stem cell international. Reproductive Biomedicine Online, 16(4), 597–598.

    Article  CAS  PubMed  Google Scholar 

  45. Delo, D. M., Olson, J., Baptista, P. M., et al. (2008). Non-invasive longitudinal tracking of human amniotic fluid stem cells in the mouse heart. Stem Cells and Development, 17(6), 1185–1194.

    Article  CAS  PubMed  Google Scholar 

  46. Sessarego, N., Parodi, A., Podesta, M., et al. (2008). Multipotent mesenchymal stromal cells from amniotic fluid: solid perspectives for clinical application. Haematologica, 93(3), 339–346.

    Article  PubMed  Google Scholar 

  47. Steigman, S. A., Armant, M., Bayer-Zwirello, L., et al. (2008). Preclinical regulatory validation of a 3-stage amniotic mesenchymal stem cell manufacturing protocol. Journal of Pediatric Surgery, 43(6), 1164–1169.

    Article  PubMed  Google Scholar 

  48. Grisafi, D., Piccoli, M., Pozzobon, M., et al. (2008). High transduction efficiency of human amniotic fluid stem cells mediated by adenovirus vectors. Stem Cells and Development, 17(5), 953–962.

    Article  CAS  PubMed  Google Scholar 

  49. Li, C., Zhou, J., Shi, G., et al. (2009). Pluripotency can be rapidly and efficiently induced in human amniotic fluid-derived cells. Human Molecular Genetics, 18(22), 4340–4349.

    Article  CAS  PubMed  Google Scholar 

  50. Ausoni, S., & Sartore, S. (2009). The cardiovascular unit as a dynamic player in disease and regeneration. Trends in Molecular Medicine, 15(12), 543–552.

    Article  PubMed  Google Scholar 

  51. Rangappa, S., Entwistle, J. W., Wechsler, A. S., & Kresh, J. Y. (2003). Cardiomyocyte-mediated contact programs human mesenchymal stem cells to express cardiogenic phenotype. The Journal of Thoracic and Cardiovascular Surgery, 126(1), 124–132.

    Article  CAS  PubMed  Google Scholar 

  52. Park, J., Setter, V., Wixler, V., & Schneider, H. (2009). Umbilical cord blood stem cells: induction of differentiation into mesenchymal lineages by cell-cell contacts with various mesenchymal cells. Tissue Engineering. Part A, 15(2), 397–406.

    Article  CAS  PubMed  Google Scholar 

  53. Choi, Y. S., Dusting, G. J., Stubbs, S., et al. (2010). Differentiation of human adipose-derived stem cells into beating cardiomyocytes. Journal of Cellular and Molecular Medicine, 14(4), 878–889.

    Article  CAS  PubMed  Google Scholar 

  54. Iijima, Y., Nagai, T., Mizukami, M., et al. (2003). Beating is necessary for transdifferentiation of skeletal muscle-derived cells into cardiomyocytes. The FASEB Journal, 17(10), 1361–1363.

    CAS  PubMed  Google Scholar 

  55. Zhu, Y., Liu, T., Song, K., Ning, R., Ma, X., & Cui, Z. (2009). ADSCs differentiated into cardiomyocytes in cardiac microenvironment. Molecular and Cellular Biochemistry, 324(1–2), 117–129.

    Article  CAS  PubMed  Google Scholar 

  56. Ishikawa, F., Shimazu, H., Shultz, L. D., et al. (2006). Purified human hematopoietic stem cells contribute to the generation of cardiomyocytes through cell fusion. The FASEB Journal, 20(7), 950–952.

    Article  CAS  PubMed  Google Scholar 

  57. Nygren, J. M., Jovinge, S., Breitbach, M., et al. (2004). Bone marrow-derived hematopoietic cells generate cardiomyocytes at a low frequency through cell fusion, but not transdifferentiation. Natural Medicines, 10(5), 494–501.

    Article  CAS  Google Scholar 

  58. Nassiri, S. M., Khaki, Z., Soleimani, M., et al. (2007). The similar effect of transplantation of marrow-derived mesenchymal stem cells with or without prior differentiation induction in experimental myocardial infarction. Journal of Biomedical Science, 14(6), 745–755.

    Article  PubMed  Google Scholar 

  59. Mazo, M., Planat-Benard, V., Abizanda, G., et al. (2008). Transplantation of adipose derived stromal cells is associated with functional improvement in a rat model of chronic myocardial infarction. European Journal of Heart Failure, 10(5), 454–462.

    Article  PubMed  Google Scholar 

  60. Yeh, Y. C., Wei, H. J., Lee, W. Y., et al. (2010). Cellular cardiomyoplasty with human amniotic fluid stem cells: in vitro and in vivo studies. Tissue Engineering. Part A, 16(6), 1925–1936.

    Article  CAS  PubMed  Google Scholar 

  61. Giebel, S., Dziaczkowska, J., Wojnar, J., et al. (2005). The impact of immunosuppressive therapy on an early quantitative NK cell reconstitution after allogeneic haematopoietic cell transplantation. Annals of Transplantation, 10(2), 29–33.

    PubMed  Google Scholar 

  62. Nifontova, I., Svinareva, D., Petrova, T., & Drize, N. (2008). Sensitivity of mesenchymal stem cells and their progeny to medicines used for the treatment of hematoproliferative diseases. Acta Haematologica, 119(2), 98–103.

    Article  PubMed  Google Scholar 

  63. Broekema, M., Harmsen, M. C., Koerts, J. A., et al. (2009). Ciclosporin does not influence bone marrow-derived cell differentiation to myofibroblasts early after renal ischemia/reperfusion. American Journal of Nephrology, 30(1), 73–83.

    Article  CAS  PubMed  Google Scholar 

  64. Terrovitis, J., Stuber, M., Youssef, A., et al. (2008). Magnetic resonance imaging overestimates ferumoxide-labeled stem cell survival after transplantation in the heart. Circulation, 117(12), 1555–1562.

    Article  PubMed  Google Scholar 

  65. Eixarch, H., Gomez, A., Kadar, E., et al. (2009). Transgene expression levels determine the immunogenicity of transduced hematopoietic grafts in partially myeloablated mice. Molecular Therapy, 17(11), 1904–1909.

    Article  CAS  PubMed  Google Scholar 

  66. Moloney, T. C., Dockery, P., Windebank, A. J., Barry, F. P., Howard, L., Dowd, E. (2010). Survival and Immunogenicity of Mesenchymal Stem Cells From the Green Fluorescent Protein Transgenic Rat in the Adult Rat Brain. Neurorehabil Neural Repair.

  67. Delo, D. M., Guan, X., Wang, Z., et al. (2010). Calcification after myocardial infarction is independent of amniotic fluid stem cell injection. Cardiovasc Pathol.

  68. Drukker, M., Katz, G., Urbach, A., et al. (2002). Characterization of the expression of MHC proteins in human embryonic stem cells. Proceedings of the National Academy of Sciences of the United States of America, 99(15), 9864–9869.

    Article  CAS  PubMed  Google Scholar 

  69. Magliocca, J. F., Held, I. K., & Odorico, J. S. (2006). Undifferentiated murine embryonic stem cells cannot induce portal tolerance but may possess immune privilege secondary to reduced major histocompatibility complex antigen expression. Stem Cells and Development, 15(5), 707–717.

    Article  CAS  PubMed  Google Scholar 

  70. Tian, L., Catt, J. W., O’Neill, C., & King, N. J. (1997). Expression of immunoglobulin superfamily cell adhesion molecules on murine embryonic stem cells. Biology of Reproduction, 57(3), 561–568.

    Article  CAS  PubMed  Google Scholar 

  71. Lampton, P. W., Crooker, R. J., Newmark, J. A., & Warner, C. M. (2008). Expression of major histocompatibility complex class I proteins and their antigen processing chaperones in mouse embryonic stem cells from fertilized and parthenogenetic embryos. Tissue Antigens, 72(5), 448–457.

    Article  CAS  PubMed  Google Scholar 

  72. Tsai, M. S., Lee, J. L., Chang, Y. J., & Hwang, S. M. (2004). Isolation of human multipotent mesenchymal stem cells from second-trimester amniotic fluid using a novel two-stage culture protocol. Human Reproduction, 19(6), 1450–1456.

    Article  PubMed  Google Scholar 

  73. Portmann-Lanz, C. B., Schoeberlein, A., Huber, A., et al. (2006). Placental mesenchymal stem cells as potential autologous graft for pre- and perinatal neuroregeneration. American Journal of Obstetrics and Gynecology, 194(3), 664–673.

    Article  CAS  PubMed  Google Scholar 

  74. Ilancheran, S., Moodley, Y., & Manuelpillai, U. (2009). Human fetal membranes: a source of stem cells for tissue regeneration and repair? Placenta, 30(1), 2–10.

    Article  CAS  PubMed  Google Scholar 

  75. Swijnenburg, R. J., Tanaka, M., Vogel, H., et al. (2005). Embryonic stem cell immunogenicity increases upon differentiation after transplantation into ischemic myocardium. Circulation, 112(9 Suppl), I166–I172.

    PubMed  Google Scholar 

  76. Dressel, R., Nolte, J., Elsner, L., et al. (2010). Pluripotent stem cells are highly susceptible targets for syngeneic, allogeneic, and xenogeneic natural killer cells. The FASEB Journal, 24(7), 2164–2177.

    Article  CAS  PubMed  Google Scholar 

  77. Dressel, R., Guan, K., Nolte, J., et al. (2009). Multipotent adult germ-line stem cells, like other pluripotent stem cells, can be killed by cytotoxic T lymphocytes despite low expression of major histocompatibility complex class I molecules. Biology Direct, 4, 31.

    Article  PubMed  Google Scholar 

  78. Pozzobon, M., Ghionzoli, M., & DeCoppi, P. (2010). ES, iPS, MSC, and AFS cells. Stem cells exploitation for Pediatric Surgery: current research and perspective. Pediatric Surgery International, 26(1), 3–10.

    Article  PubMed  Google Scholar 

  79. Cananzi, M., Atala, A., & DeCoppi, P. (2009). Stem cells derived from amniotic fluid: new potentials in regenerative medicine. Reproductive Biomedicine Online, 18(Suppl 1), 17–27.

    Article  PubMed  Google Scholar 

Download references

Acknowledgments

This work was supported by grant # 07/02 from “Città della Speranza”, Malo, Vicenza, Italy (SB, PDC) and by the Wellcome Trust (MN and AT). The authors also acknowledge the support of the Biotechnology and Biological Sciences Research Council, the British Heart Foundation and the Engineering and Physical Sciences Research Council.

Conflict of Interest and Disclosures

None to declare.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Sveva Bollini.

Electronic supplementary material

Below is the link to the electronic supplementary material.

GFP-rAFS cell with spontaneous contractile activity in co-culture with rCMs. After 4 days of co-culture, some GFP-rAFS cells were detected in CM-enriched beating areas expressing contractile activity as detected by the video recording. (MPG 583 kb)

Supplement Figure 1

Calibration curve for Endorem particles concentration mg/ml, [c] versus R2 (1/T2), demonstrating a linear relation between iron particles concentration and T2 (as long as the T2 values are between 20 and 90 seconds). (GIF 7.23 kb)

Supplement Figure 2

Analysis of differentiation of GFP-rAFS cells by immunofluorescence and gene expression analysis after 6 and 9 days of indirect co-culture with rCMs with Transwell® Membrane Inserts and after treatment with rCMs-conditioned medium. (ac) GFP-rAFS cells after indirect co-culture with rCMs and (d–f) after rCMs-conditioned medium treatment for 9 days, showing no expression of CM-specific markers as cTnT, bar, 100 μm. (g) Gel electrophoresis of RT-PCR products of control untreated GFP-rAFS cells (control GFP-rAFS cells, lane 1), GFP-rAFS cells co-cultured with rCMs on Transwell® Membrane Inserts for 6 days (lane 2), GFP-rAFS cells treated with rCM-conditioned medium for 6 days (lane 3), GFP-rAFS cells co-cultured with rCMs on Transwell® Membrane Inserts for 9 days (lane 4), GFP-rAFS cells treated with rCM-conditioned medium for 9 days (lane 5), control rCMs (lane 6) and H2O (negative control, lane 7) for the expression of the housekeeping gene β-Actin and the cardiac genes troponin I (cTnI) and sarcomeric α-actinin (cαA). GFP-rAFS cells co-cultured with rCMs with inserts and treated with rCMs-conditioned medium did not show any expression of cardiomyocyte genes (lane 2–5) compared to control undifferentiated GFP-rAFS cells (lane 1). (JPEG 2.09 mb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Bollini, S., Pozzobon, M., Nobles, M. et al. In Vitro and In Vivo Cardiomyogenic Differentiation of Amniotic Fluid Stem Cells. Stem Cell Rev and Rep 7, 364–380 (2011). https://doi.org/10.1007/s12015-010-9200-z

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12015-010-9200-z

Keywords

Navigation