DOI QR코드

DOI QR Code

Therapeutic Use of Stem Cell Transplantation for Cell Replacement or Cytoprotective Effect of Microvesicle Released from Mesenchymal Stem Cell

  • Choi, Moonhwan (Department of Bioengineering, College of Engineering, Hanyang University) ;
  • Ban, Taehyun (Department of Bioengineering, College of Engineering, Hanyang University) ;
  • Rhim, Taiyoun (Department of Bioengineering, College of Engineering, Hanyang University)
  • Received : 2013.10.30
  • Accepted : 2013.12.13
  • Published : 2014.02.28

Abstract

Idiopathic pulmonary fibrosis (IPF) is the most common and severe type of idiopathic interstitial pneumonias (IIP), and which is currently no method was developed to restore normal structure and function. There are several reports on therapeutic effects of adult stem cell transplantations in animal models of pulmonary fibrosis. However, little is known about how mesenchymal stem cell (MSC) can repair the IPF. In this study, we try to provide the evidence to show that transplanted mesenchymal stem cells directly replace fibrosis with normal lung cells using IPF model mice. As results, transplanted MSC successfully integrated and differentiated into type II lung cell which express surfactant protein. In the other hand, we examine the therapeutic effects of microvesicle treatment, which were released from mesenchymal stem cells. Though the therapeutic effects of MV treatment is less than that of MSC treatment, MV treat-ment meaningfully reduced the symptom of IPF, such as collagen deposition and inflammation. These data suggest that stem cell transplantation may be an effective strategy for the treatment of pulmonary fibrosis via replacement and cytoprotective effect of microvesicle released from MSCs.

Keywords

References

  1. Akao, Y., Iio, A., Itoh, T., Noguchi, S., Itoh, Y., Ohtsuki, Y., and Naoe, T. (2011). Microvesicle-mediated RNA molecule delivery system using monocytes/macrophages. Mol. Ther. 19, 395-399. https://doi.org/10.1038/mt.2010.254
  2. Banerjee, E.R., Laflamme, M.A., Papayannopoulou, T., Kahn, M., Murry, C.E., and Henderson, W.R., Jr. (2012). Human embryonic stem cells differentiated to lung lineage-specific cells ameliorate pulmonary fibrosis in a xenograft transplant mouse model. PLoS One 7, e33165. https://doi.org/10.1371/journal.pone.0033165
  3. Bedrossian, C., Warren, C., Ohar, J., and Jackson, F. (1991). Amiodarone pulmonary toxicity - cytopathology ultrastructure and immunocytochemistry. Lab. Invest. 64, A113-A113.
  4. Camussi, G., Deregibus, M.C., Bruno, S., Cantaluppi, V., and Biancone, L. (2010). Exosomes/microvesicles as a mechanism of cell-to-cell communication. Kidney Int. 78, 838-848. https://doi.org/10.1038/ki.2010.278
  5. Guttentag, S.H., Beers, M.F., Bieler, B.M., and Ballard, P.L. (1998). Surfactant protein B processing in human fetal lung. Am. J. Physiol. 275, L559-566.
  6. Ha, Y., Shin, J.S., Lee, D.Y., and Rhim, T. (2012). Fluorescently labeled nanoparticles enable the detection of stem cell-derived hepatocytes. B Korean Chem. Soc. 33, 1983-1988. https://doi.org/10.5012/bkcs.2012.33.6.1983
  7. Hu, G., Drescher, K.M., and Chen, X.M. (2012). Exosomal miRNAs: biological properties and therapeutic potential. Front. Genet. 3, 56.
  8. Iyer, S.N., Gurujeyalakshmi, G., and Giri, S.N. (1999). Effects of pirfenidone on transforming growth factor-beta gene expression at the transcriptional level in bleomycin hamster model of lung fibrosis. J. Pharmacol. Exp. Ther. 291, 367-373.
  9. Kim, T.H., Lee, Y.H., Kim, K.H., Lee, S.H., Cha, J.Y., Shin, E.K., Jung, S., Jang, A.S., Park, S.W., Uh, S.T., et al. (2010). Role of lung apolipoprotein A-I in idiopathic pulmonary fibrosis: antiinflammatory and antifibrotic effect on experimental lung injury and fibrosis. Am. J. Respir. Crit. Care Med. 182, 633-642. https://doi.org/10.1164/rccm.200905-0659OC
  10. King, T.E., Jr., Tooze, J.A., Schwarz, M.I., Brown, K.R., and Cherniack, R.M. (2001). Predicting survival in idiopathic pulmonary fibrosis: scoring system and survival model. Am. J. Respir. Crit. Care Med. 164, 1171-1181. https://doi.org/10.1164/ajrccm.164.7.2003140
  11. Lee, S.H., Jang, A.S., Kim, Y.E., Cha, J.Y., Kim, T.H., Jung, S., Park, S.K., Lee, Y.K., Won, J.H., Kim, Y.H., et al. (2010). Modulation of cytokine and nitric oxide by mesenchymal stem cell transfer in lung injury/fibrosis. Respir. Res. 11, 16. https://doi.org/10.1186/1465-9921-11-16
  12. Liu, T.J., Chung, M.J., Ullenbruch, M., Yu, H.F., Jin, H., Hu, B., Choi, Y.Y., Ishikawa, F., and Phan, S.H. (2007). Telomerase activity is required for bleomycin-induced pulmonary fibrosis in mice. J. Clin. Invest. 117, 3800-3809.
  13. Luppi, F., Spagnolo, P., Cerri, S., and Richeldi, L. (2012). The big clinical trials in idiopathic pulmonary fibrosis. Curr. Opin. Pulm. Med. 18, 428-432. https://doi.org/10.1097/MCP.0b013e3283567ff9
  14. Moore, B.B., and Hogaboam, C.M. (2008). Murine models of pulmonary fibrosis. Am. J. Physiol. Lung C 294, L152-L160. https://doi.org/10.1152/ajplung.00313.2007
  15. Ortiz, L.A., Gambelli, F., McBride, C., Gaupp, D., Baddoo, M., Kaminski, N., and Phinney, D.G. (2003). Mesenchymal stem cell engraftment in lung is enhanced in response to bleomycin exposure and ameliorates its fibrotic effects. Proc. Natl. Acad. Sci. USA 100, 8407-8411. https://doi.org/10.1073/pnas.1432929100
  16. Raghu, G., Collard, H.R., Egan, J.J., Martinez, F.J., Behr, J., Brown, K.K., Colby, T.V., Cordier, J.F., Flaherty, K.R., Lasky, J.A., et al. (2011). An official ATS/ERS/JRS/ALAT statement: idiopathic pulmonary fibrosis: evidence-based guidelines for diagnosis and management. Am. J. Respir. Crit. Care Med. 183, 788-824. https://doi.org/10.1164/rccm.2009-040GL
  17. Selman, M., King, T.E., Pardo, A., American Thoracic, S., European Respiratory, S., and American College of Chest, P. (2001). Idiopathic pulmonary fibrosis: prevailing and evolving hypotheses about its pathogenesis and implications for therapy. Ann. Int. Med. 134, 136-151. https://doi.org/10.7326/0003-4819-134-2-200101160-00015
  18. Steele, M.P., and Schwartz, D.A. (2012). Molecular mechanisms in progressive idiopathic pulmonary fibrosis. Ann. Rev. Med. 64, 265-276.
  19. Szade, K., Zuba-Surma, E., Rutkowski, A.J., Jozkowicz, A., and Dulak, J. (2011). CD45-CD14 +CD34 + murine bone marrow low-adherent mesenchymal primitive cells preserve multilineage differentiation potential in long-term in vitro culture. Mol. Cells 31, 497-507. https://doi.org/10.1007/s10059-011-2176-y
  20. Taniguchi, H., Ebina, M., Kondoh, Y., Ogura, T., Azuma, A., Suga, M., Taguchi, Y., Takahashi, H., Nakata, K., Sato, A., et al. (2010). Pirfenidone in idiopathic pulmonary fibrosis. Eur. Respir. J. 35, 821-829. https://doi.org/10.1183/09031936.00005209
  21. Zhao, F., Zhang, Y.F., Liu, Y.G., Zhou, J.J., Li, Z.K., Wu, C.G., and Qi, H.W. (2008). Therapeutic effects of bone marrow-derived mesenchymal stem cells engraftment on bleomycin-induced lung injury in rats. Transplant. Proc. 40, 1700-1705. https://doi.org/10.1016/j.transproceed.2008.01.080
  22. Zhao, M.M., Cui, J.Z., Cui, Y., Li, R., Tian, Y.X., Song, S.X., Zhang, J., and Gao, J.L. (2013). Therapeutic effect of exogenous bone marrowderived mesenchymal stem cell transplantation on silicosis via paracrine mechanisms in rats. Mol. Med. Rep. 8, 741-746. https://doi.org/10.3892/mmr.2013.1580

Cited by

  1. From Here to There, Progenitor Cells and Stem Cells Are Everywhere in Lung Vascular Remodeling vol.4, 2016, https://doi.org/10.3389/fped.2016.00080
  2. Effect of the Microenvironment on Mesenchymal Stem Cell Paracrine Signaling: Opportunities to Engineer the Therapeutic Effect vol.26, pp.9, 2017, https://doi.org/10.1089/scd.2016.0349
  3. Mesenchymal stromal cells improve cardiac function and left ventricular remodeling in a heart transplantation model vol.34, pp.11, 2015, https://doi.org/10.1016/j.healun.2015.05.008
  4. Exosomes and Their Therapeutic Potentials of Stem Cells vol.2016, 2016, https://doi.org/10.1155/2016/7653489
  5. Mesenchymal Stromal Cells in Animal Bleomycin Pulmonary Fibrosis Models: A Systematic Review vol.4, pp.12, 2015, https://doi.org/10.5966/sctm.2015-0121
  6. Cell-Based Therapy for Silicosis vol.2016, 2016, https://doi.org/10.1155/2016/5091838
  7. Concise Review: MSC-Derived Exosomes for Cell-Free Therapy vol.35, pp.4, 2017, https://doi.org/10.1002/stem.2575
  8. Molecular signatures of mesenchymal stem cell-derived extracellular vesicle-mediated tissue repair vol.6, pp.1, 2015, https://doi.org/10.1186/s13287-015-0214-y
  9. Will stem cells bring hope to pathological skin scar treatment? vol.18, pp.8, 2016, https://doi.org/10.1016/j.jcyt.2016.05.008
  10. Oncostatin M-Preconditioned Mesenchymal Stem Cells Alleviate Bleomycin-Induced Pulmonary Fibrosis Through Paracrine Effects of the Hepatocyte Growth Factor vol.6, pp.3, 2017, https://doi.org/10.5966/sctm.2016-0054
  11. Extracellular Vesicles: Evolving Factors in Stem Cell Biology vol.2016, 2016, https://doi.org/10.1155/2016/1073140
  12. Exosome Derived From Human Umbilical Cord Mesenchymal Stem Cell Mediates MiR-181c Attenuating Burn-induced Excessive Inflammation vol.8, 2016, https://doi.org/10.1016/j.ebiom.2016.04.030
  13. Management of Fibrosis: The Mesenchymal Stromal Cells Breakthrough vol.2014, 2014, https://doi.org/10.1155/2014/340257
  14. Osteoblast-Derived Extracellular Vesicles Are Biological Tools for the Delivery of Active Molecules to Bone vol.33, pp.3, 2017, https://doi.org/10.1002/jbmr.3332
  15. Mesenchymal stem cells and cell-derived extracellular vesicles protect hippocampal neurons from oxidative stress and synapse damage induced by amyloid-β oligomers vol.293, pp.6, 2017, https://doi.org/10.1074/jbc.M117.807180
  16. Clinical Application of Mesenchymal Stem Cell-Derived Extracellular Vesicle-Based Therapeutics for Inflammatory Lung Diseases vol.7, pp.10, 2018, https://doi.org/10.3390/jcm7100355
  17. Exosomes and Their MicroRNA Cargo: New Players in Peripheral Nerve Regeneration vol.32, pp.9, 2018, https://doi.org/10.1177/1545968318798955
  18. Bioprocessing of Mesenchymal Stem Cells and Their Derivatives: Toward Cell-Free Therapeutics vol.2018, pp.1687-9678, 2018, https://doi.org/10.1155/2018/9415367
  19. NMDA receptor activation inhibits the antifibrotic effect of BM-MSCs on bleomycin-induced pulmonary fibrosis vol.315, pp.3, 2018, https://doi.org/10.1152/ajplung.00002.2018
  20. Mesenchymal Stem Cell Therapy of Pulmonary Fibrosis pp.1555-3892, 2018, https://doi.org/10.1177/0963689718787501
  21. Extracellular Vesicles Derived from Hypoxic Human Mesenchymal Stem Cells Attenuate GSK3β Expression via miRNA-26a in an Ischemia-Reperfusion Injury Model vol.59, pp.6, 2018, https://doi.org/10.3349/ymj.2018.59.6.736
  22. Mesenchymal Stem Cells on Horizon: A New Arsenal of Therapeutic Agents vol.14, pp.4, 2018, https://doi.org/10.1007/s12015-018-9817-x
  23. Human mesenchymal stem cell-derived extracellular vesicles/estrogen combined therapy safely ameliorates experimentally induced intrauterine adhesions in a female rat model vol.9, pp.1, 2018, https://doi.org/10.1186/s13287-018-0924-z
  24. Novel aspects of pathogenesis and regeneration mechanisms in COPD vol.10, pp.None, 2015, https://doi.org/10.2147/copd.s82518
  25. Mesenchymal stem cells in idiopathic pulmonary fibrosis vol.8, pp.60, 2014, https://doi.org/10.18632/oncotarget.18126
  26. Therapeutic potential of products derived from mesenchymal stem/stromal cells in pulmonary disease vol.19, pp.None, 2014, https://doi.org/10.1186/s12931-018-0921-x
  27. Emerging Role of Mesenchymal Stem Cell-derived Exosomes in Regenerative Medicine vol.14, pp.6, 2014, https://doi.org/10.2174/1574888x14666190228103230
  28. Prospective application of stem cells to prevent post‐operative skeletal fibrosis vol.37, pp.6, 2014, https://doi.org/10.1002/jor.24266
  29. Mesenchymal Stem Cells in Homeostasis and Systemic Diseases: Hypothesis, Evidences, and Therapeutic Opportunities vol.20, pp.15, 2014, https://doi.org/10.3390/ijms20153738
  30. Exosomes: Carriers of Pro-Fibrotic Signals and Therapeutic Targets in Fibrosis vol.25, pp.42, 2014, https://doi.org/10.2174/1381612825666191209161443
  31. The potential of mesenchymal stem cell therapy for chronic lung disease vol.14, pp.1, 2014, https://doi.org/10.1080/17476348.2020.1679628
  32. Effects of Mesenchymal Stem Cell Coculture on Human Lung Small Airway Epithelial Cells vol.2020, pp.None, 2014, https://doi.org/10.1155/2020/9847579
  33. Therapeutic Potential of Exosomes in Pulmonary Fibrosis vol.11, pp.None, 2014, https://doi.org/10.3389/fphar.2020.590972
  34. Therapeutic potential of mesenchymal stem/stromal cell-derived secretome and vesicles for lung injury and disease vol.20, pp.2, 2020, https://doi.org/10.1080/14712598.2020.1689954
  35. Extracellular vesicles and asthma: A review of the literature vol.50, pp.3, 2020, https://doi.org/10.1111/cea.13562
  36. Therapeutic Use of Extracellular Vesicles for Acute and Chronic Lung Disease vol.21, pp.7, 2020, https://doi.org/10.3390/ijms21072318
  37. Exosomes as mediators and biomarkers in fibrosis vol.14, pp.8, 2020, https://doi.org/10.2217/bmm-2019-0368
  38. Mesenchymal Stem Cell (MSC)‐Derived Extracellular Vesicles: Potential Therapeutics as MSC Trophic Mediators in Regenerative Medicine vol.303, pp.6, 2014, https://doi.org/10.1002/ar.24186
  39. Emerging role of extracellular vesicles in the respiratory system vol.52, pp.6, 2014, https://doi.org/10.1038/s12276-020-0450-9
  40. Neural Stem Cell-Derived Exosomes Revert HFD-Dependent Memory Impairment via CREB-BDNF Signalling vol.21, pp.23, 2014, https://doi.org/10.3390/ijms21238994
  41. The development of mesenchymal stem cell therapy in the present, and the perspective of cell-free therapy in the future vol.27, pp.1, 2014, https://doi.org/10.3350/cmh.2020.0194
  42. Recent Findings on Cell-Based Therapies for COVID19-Related Pulmonary Fibrosis vol.30, pp.None, 2021, https://doi.org/10.1177/0963689721996217
  43. The Current Status of Mesenchymal Stromal Cells: Controversies, Unresolved Issues and Some Promising Solutions to Improve Their Therapeutic Efficacy vol.9, pp.None, 2014, https://doi.org/10.3389/fcell.2021.650664
  44. Effects of Mesenchymal Stromal Cell-Derived Extracellular Vesicles in Lung Diseases: Current Status and Future Perspectives vol.17, pp.2, 2014, https://doi.org/10.1007/s12015-020-10085-8
  45. New Insights into Pathomechanisms and Treatment Possibilities for Lung Silicosis vol.22, pp.8, 2014, https://doi.org/10.3390/ijms22084162
  46. Mesenchymal Stromal Cell Secretome for Post-COVID-19 Pulmonary Fibrosis: A New Therapy to Treat the Long-Term Lung Sequelae? vol.10, pp.5, 2014, https://doi.org/10.3390/cells10051203
  47. Mesenchymal stem cells ameliorate silica‐induced pulmonary fibrosis by inhibition of inflammation and epithelial‐mesenchymal transition vol.25, pp.13, 2014, https://doi.org/10.1111/jcmm.16621
  48. Mesenchymal stromal cell extracellular vesicles as therapy for acute and chronic respiratory diseases: A meta‐analysis vol.10, pp.12, 2014, https://doi.org/10.1002/jev2.12141
  49. Characterizing Organelles in Live Stem Cells Using Label-Free Optical Diffraction Tomography vol.44, pp.11, 2014, https://doi.org/10.14348/molcells.2021.0190