Skip to main content
Log in

Transplantation of PSA-NCAM-Positive Neural Precursors from Human Embryonic Stem Cells Promotes Functional Recovery in an Animal Model of Spinal Cord Injury

  • Original Article
  • Published:
Tissue Engineering and Regenerative Medicine Aims and scope

A Correction to this article was published on 26 October 2022

This article has been updated

Abstract

Background:

Spinal cord injury (SCI) results in permanent impairment of motor and sensory functions at and below the lesion site. There is no therapeutic option to the functional recovery of SCI involving diverse injury responses of different cell types in the lesion that limit endogenous nerve regeneration. In this regard, cell replacement therapy utilizing stem cells or their derivatives has become a highly promising approach to promote locomotor recovery. For this reason, the demand for a safe and efficient multipotent cell source that can differentiate into various neural cells is increasing. In this study, we evaluated the efficacy and safety of human polysialylated-neural cell adhesion molecule (PSA-NCAM)-positive neural precursor cells (hNPCsPSA-NCAM+) as a treatment for SCI.

Methods:

One hundred thousand hNPCsPSA-NCAM+ isolated from human embryonic stem cell-derived NPCs were transplanted into the lesion site by microinjection 7 days after contusive SCI at the thoracic level. We examined the histological characteristics of the graft and behavioral improvement in the SCI rats 10 weeks after transplantation.

Results:

Locomotor activity improvement was estimated by the Basso–Beattie–Bresnahan locomotor rating scale. Behavioral tests revealed that the transplantation of the hNPCsPSA-NCAM+ into the injured spinal cords of rats significantly improved locomotor function. Histological examination showed that hNPCsPSA-NCAM+ had differentiated into neural cells and successfully integrated into the host tissue with no evidence of tumor formation. We investigated cytokine expressions, which led to the early therapeutic effect of hNPCsPSA-NCAM+, and found that some undifferentiated NPCs still expressed midkine, a well-known neurotrophic factor involved in neural development and inflammatory responses, 10 weeks after transplantation.

Conclusion:

Our results demonstrate that hNPCsPSA-NCAM+ serve as a safe and efficient cell source which has the potential to improve impaired motor function following SCI.

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.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5

Similar content being viewed by others

Change history

References

  1. Noble M, Mayer-Pröschel M, Davies JE, Davies SJ, Pröschel C. Cell therapies for the central nervous system: How do we identify the best candidates? Curr Opin Neurol. 2011;24:570–6.

    PubMed  Google Scholar 

  2. Fawcett JW, Asher RA. The glial scar and central nervous system repair. Brain Res Bull. 1999;49:377–91.

    PubMed  CAS  Google Scholar 

  3. Parr AM, Tator CH, Keating A. Bone marrow-derived mesenchymal stromal cells for the repair of central nervous system injury. Bone Marrow Transplant. 2007;40:609–19.

    PubMed  CAS  Google Scholar 

  4. Tetzlaff W, Okon EB, Karimi-Abdolrezaee S, Hill CE, Sparling JS, Plemel JR, et al. A systematic review of cellular transplantation therapies for spinal cord injury. J Neurotrauma. 2011;28:1611–82.

    PubMed  PubMed Central  Google Scholar 

  5. Moreno-Manzano V, Rodríguez-Jiménez FJ, García-Roselló M, Laínez S, Erceg S, Calvo MT, et al. Activated spinal cord ependymal stem cells rescue neurological function. Stem Cells. 2009;27:733–43.

    PubMed  Google Scholar 

  6. McDonald JW, Liu XZ, Qu Y, Liu S, Mickey SK, Turetsky D, et al. Transplanted embryonic stem cells survive, differentiate and promote recovery in injured rat spinal cord. Nat Med. 1999;5:1410–2.

    PubMed  CAS  Google Scholar 

  7. Keirstead HS, Nistor G, Bernal G, Totoiu M, Cloutier F, Sharp K, et al. Human embryonic stem cell-derived oligodendrocyte progenitor cell transplants remyelinate and restore locomotion after spinal cord injury. J Neurosci. 2005;25:4694–705.

    PubMed  PubMed Central  CAS  Google Scholar 

  8. Kim DS, Jung SJ, Lee JS, Lim BY, Kim HA, Yoo JE, et al. Rapid generation of OPC-like cells from human pluripotent stem cells for treating spinal cord injury. Exp Mol Med. 2017;49:e361.

    PubMed  PubMed Central  Google Scholar 

  9. Mothe AJ, Tator CH. Advances in stem cell therapy for spinal cord injury. J Clin Invest. 2012;122:3824–34.

    PubMed  PubMed Central  CAS  Google Scholar 

  10. Zhang SC, Wernig M, Duncan ID, Brüstle O, Thomson JA. In vitro differentiation of transplantable neural precursors from human embryonic stem cells. Nat Biotechnol. 2001;19:1129–33.

    PubMed  CAS  Google Scholar 

  11. Elkabetz Y, Panagiotakos G, Al Shamy G, Socci ND, Tabar V, Studer L. Human ES cell-derived neural rosettes reveal a functionally distinct early neural stem cell stage. Genes Dev. 2008;22:152–65.

    PubMed  PubMed Central  CAS  Google Scholar 

  12. Kim DS, Lee JS, Leem JW, Huh YJ, Kim JY, Kim HS, et al. Robust enhancement of neural differentiation from human ES and iPS cells regardless of their innate difference in differentiation propensity. Stem Cell Rev Rep. 2010;6:270–81.

    PubMed  CAS  Google Scholar 

  13. Arnhold S, Klein H, Semkova I, Addicks K, Schraermeyer U. Neurally selected embryonic stem cells induce tumor formation after long-term survival following engraftment into the subretinal space. Invest Ophthalmol Vis Sci. 2004;45:4251–5.

    PubMed  Google Scholar 

  14. Doi D, Morizane A, Kikuchi T, Onoe H, Hayashi T, Kawasaki T, et al. Prolonged maturation culture favors a reduction in the tumorigenicity and the dopaminergic function of human ESC-derived neural cells in a primate model of Parkinson's disease. Stem Cells. 2012;30:935–45.

  15. Seminatore C, Polentes J, Ellman D, Kozubenko N, Itier V, Tine S, et al. The postischemic environment differentially impacts teratoma or tumor formation after transplantation of human embryonic stem cell-derived neural progenitors. Stroke. 2010;41:153–9.

    PubMed  Google Scholar 

  16. Kim DS, Lee DR, Kim HS, Yoo JE, Jung SJ, Lim BY, et al. Highly pure and expandable PSA-NCAM-positive neural precursors from human ESC and iPSC-derived neural rosettes. PLoS One. 2012;7:e39715.

    PubMed  PubMed Central  CAS  Google Scholar 

  17. Lee DR, Yoo JE, Lee JS, Park S, Lee J, Park CY, et al. PSA-NCAM-negative neural crest cells emerging during neural induction of pluripotent stem cells cause mesodermal tumors and unwanted grafts. Stem Cell Reports. 2015;4:821–34.

    PubMed  PubMed Central  CAS  Google Scholar 

  18. Jang J, Yoo JE, Lee JA, Lee DR, Kim JY, Huh YJ, et al. Disease-specific induced pluripotent stem cells: a platform for human disease modeling and drug discovery. Exp Mol Med. 2012;44:202–13.

    PubMed  CAS  Google Scholar 

  19. Basso DM, Beattie MS, Bresnahan JC. Graded histological and locomotor outcomes after spinal cord contusion using the NYU weight-drop device versus transection. Exp Neurol. 1996;139:244–56.

    PubMed  CAS  Google Scholar 

  20. Willis CM, Nicaise AM, Peruzzotti-Jametti L, Pluchino S. The neural stem cell secretome and its role in brain repair. Brain Res. 2020;1729:146615.

    PubMed  Google Scholar 

  21. Yang H, Wang C, Chen H, Li L, Ma S, Wang H, et al. Neural stem cell-conditioned medium ameliorated cerebral ischemia-reperfusion injury in rats. Stem Cells Int. 2018;2018:4659159.

    PubMed  PubMed Central  Google Scholar 

  22. Červenka J, Tylečková J, Kupcová Skalníková H, Vodičková Kepková K, Poliakh I, Valeková I, et al. Proteomic characterization of human neural stem cells and their secretome during in vitro differentiation. Front Cell Neurosci. 2021;14:612560.

    PubMed  PubMed Central  Google Scholar 

  23. Weckbach LT, Muramatsu T, Walzog B. Midkine in inflammation. ScientificWorldJournal. 2011;11:2491–505.

    PubMed  PubMed Central  Google Scholar 

  24. Winkler C, Yao S. The midkine family of growth factors: diverse roles in nervous system formation and maintenance. Br J Pharmacol. 2014;171:905–12.

    PubMed  PubMed Central  CAS  Google Scholar 

  25. Yoshida Y, Sakakima H, Matsuda F, Ikutomo M. Midkine in repair of the injured nervous system. Br J Pharmacol. 2014;171:924–30.

    PubMed  PubMed Central  CAS  Google Scholar 

  26. Sakakima H, Yoshida Y, Muramatsu T, Yone K, Goto M, Ijiri K, et al. Traumatic injury-induced midkine expression in the adult rat spinal cord during the early stage. J Neurotrauma. 2004;21:471–7.

    PubMed  Google Scholar 

  27. Muramoto A, Imagama S, Natori T, Wakao N, Ando K, Tauchi R, et al. Midkine overcomes neurite outgrowth inhibition of chondroitin sulfate proteoglycan without glial activation and promotes functional recovery after spinal cord injury. Neurosci Lett. 2013;550:150–5.

    PubMed  CAS  Google Scholar 

  28. Yoshida Y, Goto M, Tsutsui J, Ozawa M, Sato E, Osame M, et al. Midkine is present in the early stage of cerebral infarct. Brain Res Dev Brain Res. 1995;85:25–30.

    PubMed  CAS  Google Scholar 

  29. Assinck P, Duncan GJ, Hilton BJ, Plemel JR, Tetzlaff W. Cell transplantation therapy for spinal cord injury. Nat Neurosci. 2017;20:637–47.

    PubMed  CAS  Google Scholar 

  30. Lu P, Wang Y, Graham L, McHale K, Gao M, Wu D, et al. Long-distance growth and connectivity of neural stem cells after severe spinal cord injury. Cell. 2012;150:1264–73.

    PubMed  PubMed Central  CAS  Google Scholar 

  31. Kadoya K, Lu P, Nguyen K, Lee-Kubli C, Kumamaru H, Yao L, et al. Spinal cord reconstitution with homologous neural grafts enables robust corticospinal regeneration. Nat Med. 2016;22:479–87.

    PubMed  PubMed Central  CAS  Google Scholar 

  32. Lu P, Woodruff G, Wang Y, Graham L, Hunt M, Wu D, et al. Long-distance axonal growth from human induced pluripotent stem cells after spinal cord injury. Neuron. 2014;83:789–96.

    PubMed  PubMed Central  CAS  Google Scholar 

  33. He Z, Jin Y. Intrinsic control of axon regeneration. Neuron. 2016;90:437–51.

    PubMed  CAS  Google Scholar 

  34. Kakulas BA. Neuropathology: the foundation for new treatments in spinal cord injury. Spinal Cord. 2004;42:549–63.

    PubMed  CAS  Google Scholar 

  35. Crowe MJ, Bresnahan JC, Shuman SL, Masters JN, Beattie MS. Apoptosis and delayed degeneration after spinal cord injury in rats and monkeys. Nat Med. 1997;3:73–6.

    PubMed  CAS  Google Scholar 

  36. Plemel JR, Keough MB, Duncan GJ, Sparling JS, Yong VW, Stys PK, et al. Remyelination after spinal cord injury: is it a target for repair? Prog Neurobiol. 2014;117:54–72.

    PubMed  CAS  Google Scholar 

  37. Cummings BJ, Uchida N, Tamaki SJ, Salazar DL, Hooshmand M, Summers R, et al. Human neural stem cells differentiate and promote locomotor recovery in spinal cord-injured mice. Proc Natl Acad Sci USA. 2005;102:14069–74.

    PubMed  PubMed Central  CAS  Google Scholar 

  38. Barnabé-Heider F, Frisén J. Stem cells for spinal cord repair. Cell Stem Cell. 2008;3:16–24.

    PubMed  Google Scholar 

  39. Kumamaru H, Ohkawa Y, Saiwai H, Yamada H, Kubota K, Kobayakawa K, et al. Direct isolation and RNA-seq reveal environment-dependent properties of engrafted neural stem/progenitor cells. Nat Commun. 2012;3:1140.

    PubMed  Google Scholar 

  40. Nishimura S, Yasuda A, Iwai H, Takano M, Kobayashi Y, Nori S, et al. Time-dependent changes in the microenvironment of injured spinal cord affects the therapeutic potential of neural stem cell transplantation for spinal cord injury. Mol Brain. 2013;6:3.

    PubMed  PubMed Central  Google Scholar 

  41. Kadomatsu K, Huang RP, Suganuma T, Murata F, Muramatsu T. A retinoic acid responsive gene MK found in the teratocarcinoma system is expressed in spatially and temporally controlled manner during mouse embryogenesis. J Cell Biol. 1990;110:607–16.

    PubMed  CAS  Google Scholar 

  42. Fan QW, Muramatsu T, Kadomatsu K. Distinct expression of midkine and pleiotrophin in the spinal cord and placental tissues during early mouse development. Dev Growth Differ. 2000;42:113–9.

    PubMed  CAS  Google Scholar 

  43. Muramatsu H, Muramatsu T. Purification of recombinant midkine and examination of its biological activities: functional comparison of new heparin binding factors. Biochem Biophys Res Commun. 1991;177:652–8.

    PubMed  CAS  Google Scholar 

  44. Muramatsu H, Shirahama H, Yonezawa S, Maruta H, Muramatsu T. Midkine, a retinoic acid-inducible growth/differentiation factor: immunochemical evidence for the function and distribution. Dev Biol. 1993;159:392–402.

    PubMed  CAS  Google Scholar 

  45. Michikawa M, Kikuchi S, Muramatsu H, Muramatsu T, Kim SU. Retinoic acid responsive gene product, midkine, has neurotrophic functions for mouse spinal cord and dorsal root ganglion neurons in culture. J Neurosci Res. 1993;35:530–9.

    PubMed  CAS  Google Scholar 

Download references

Acknowledgements

We thank Mrs. S.J. Jung for technical assistance. This work was supported by the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare, Republic of Korea (HI20C0168).

Author information

Authors and Affiliations

Authors

Contributions

D.-H. Kim Ph.D.: Conception and design, Collection and/or assembly of data, Data analysis and interpretation, Manuscript writing. H.-J. Cho MS: Collection and/or assembly of data, Data analysis and interpretation. C.-Y. Park Ph.D.: Collection and/or assembly of data, Data analysis and interpretation. M.S. Cho Ph.D.: Conception and design, Data analysis and interpretation, Manuscript writing and Final approval of manuscript. D.-W. Kim Ph.D.: Conception and design, Data analysis and interpretation, Manuscript writing and Final approval of manuscript.

Corresponding authors

Correspondence to Myung Soo Cho or Dong-Wook Kim.

Ethics declarations

Conflict of interest

The authors have no conflicts of interest to declare.

Ethical statement

All animal studies were done with the approval of the Institutional Animal Care and Use Committee of Yonsei University College of Medicine, Seoul, Korea (4-2015-1097).

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Springer Nature or its licensor holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kim, DH., Cho, HJ., Park, CY. et al. Transplantation of PSA-NCAM-Positive Neural Precursors from Human Embryonic Stem Cells Promotes Functional Recovery in an Animal Model of Spinal Cord Injury. Tissue Eng Regen Med 19, 1349–1358 (2022). https://doi.org/10.1007/s13770-022-00483-z

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s13770-022-00483-z

Keywords

Navigation