Generic placeholder image

Current Stem Cell Research & Therapy

Editor-in-Chief

ISSN (Print): 1574-888X
ISSN (Online): 2212-3946

Review Article

The Application of Neural Stem/Progenitor Cells for Regenerative Therapy of Spinal Cord Injury

Author(s): Chao Yu, Kaishun Xia, Zhe Gong, Liwei Ying, Jiawei Shu, Feng Zhang, Qixin Chen*, Fangcai Li* and Chengzhen Liang*

Volume 14, Issue 6, 2019

Page: [495 - 503] Pages: 9

DOI: 10.2174/1574888X14666190329095638

Price: $65

Abstract

Spinal cord injury (SCI) is a devastating event, and there are still no effective therapies currently available. Neural stem cells (NSCs) have gained increasing attention as promising regenerative therapy of SCI. NSCs based therapies of various neural diseases in animal models and clinical trials have been widely investigated. In this review we aim to summarize the development and recent progress in the application of NSCs in cell transplantation therapy for SCI. After brief introduction on sequential genetic steps regulating spinal cord development in vivo, we describe current experimental approaches for neural induction of NSCs in vitro. In particular, we focus on NSCs induced from pluripotent stem cells (PSCs). Finally, we highlight recent progress on the NSCs, which show great promise in the application to regeneration therapy for SCI.

Keywords: Spinal cord injury, neural induction, neural stem cell, pluripotent stem cell, patient, macrophages.

[1]
Jazayeri S, Beygi S, Shokraneh F, Hagen E, Rahimi-Movaghar V. Incidence of traumatic spinal cord injury worldwide: A systematic review. Eur Spine J 2015; 24: 905-18.
[2]
Salewski R, Mitchell R, Li L, et al. Transplantation of induced pluripotent stem cell-derived neural stem cells mediate functional recovery following thoracic spinal cord injury through remyelination of axons. Stem Cells Transl Med 2015; 4: 743-54.
[3]
Lu P, Wang Y, Graham L, et al. Long-distance growth and connectivity of neural stem cells after severe spinal cord injury. Cell 2012; 150: 1264-73.
[4]
Kadoya K, Lu P, Nguyen K, et al. Spinal cord reconstitution with homologous neural grafts enables robust corticospinal regeneration. Nature medicine 2016; 22: 479-87.
[5]
Bunge R, Puckett W, Becerra J, Marcillo A, Quencer R. Observations on the pathology of human spinal cord injury. A review and classification of 22 new cases with details from a case of chronic cord compression with extensive focal demyelination. Adv Neurol 1993; 59: 75-89.
[6]
Oyinbo C. Secondary injury mechanisms in traumatic spinal cord injury: a nugget of this multiply cascade. Acta Neurobiol Exp (Warsz) 2011; 71: 281-99.
[7]
Assinck P, Duncan GJ, Hilton BJ, Plemel JR, Tetzlaff W. Cell transplantation therapy for spinal cord injury. Nat Neurosci 2017; 20: 637-47.
[8]
Filbin M. Myelin-associated inhibitors of axonal regeneration in the adult mammalian CNS. Nat Rev Neurosci 2003; 4: 703-13.
[9]
Frontini-López YRGA, Masone D, Bustos DM, Uhart M. Adipose-derived mesenchymal stem/stromal cells: From the lab bench to the basic concepts for clinical translation. Biocell 2018; 42: 67-77.
[10]
Yasuda A, Tsuji O, Shibata S, et al. Significance of remyelination by neural stem/progenitor cells transplanted into the injured spinal cord. Stem Cells 2011; 29: 1983-94.
[11]
Hawryluk G, Mothe A, Wang J, Wang S, Tator C, Fehlings M. An in vivo characterization of trophic factor production following neural precursor cell or bone marrow stromal cell transplantation for spinal cord injury. Stem Cells Dev 2012; 21: 2222-38.
[12]
Gage F. Mammalian neural stem cells. Science 2000; 287: 1433-8.
[13]
Nagoshi N, Okano H. iPSC-derived neural precursor cells: Potential for cell transplantation therapy in spinal cord injury. Cell Mol Life Sci 2018; 75: 989-1000.
[14]
Zhao T, Zhang ZN, Rong Z, Xu Y. Immunogenicity of induced pluripotent stem cells. Nature 2011; 474: 212-5.
[15]
Itakura G, Ozaki M, Nagoshi N, et al. Low immunogenicity of mouse induced pluripotent stem cell-derived neural stem/progenitor cells. Scientific reports 2017; 7: 12996.
[16]
Le Dréau G, Martí E. Dorsal-ventral patterning of the neural tube: A tale of three signals. Dev Neurobiol 2012; 72: 1471-81.
[17]
Munoz EM. Microglia-precursor cell interactions in health and in pathology. Biocell 2018; 42: 1-5.
[18]
Jessell T. Neuronal specification in the spinal cord: Inductive signals and transcriptional codes. Nat Rev Genet 2000; 1: 20-9.
[19]
Briscoe J, Pierani A, Jessell T, Ericson J. A homeodomain protein code specifies progenitor cell identity and neuronal fate in the ventral neural tube. Cell 2000; 101: 435-45.
[20]
Panman L, Andersson E, Alekseenko Z, et al. Transcription factor-induced lineage selection of stem-cell-derived neural progenitor cells. Cell Stem Cell 2011; 8: 663-75.
[21]
Mazzoni EO, Mahony S, Peljto M, et al. Saltatory remodeling of Hox chromatin in response to rostrocaudal patterning signals. Nat Neurosci 2013; 16: 1191-8.
[22]
Lippmann E, Williams C, Ruhl D, et al. Deterministic HOX patterning in human pluripotent stem cell-derived neuroectoderm. Stem Cell Reports 2015; 4: 632-44.
[23]
McDonald J, Liu X, Qu Y, et al. Transplanted embryonic stem cells survive, differentiate and promote recovery in injured rat spinal cord. Nat Med 1999; 5: 1410-2.
[24]
Thier M, Wörsdörfer P, Lakes Y, et al. Direct conversion of fibroblasts into stably expandable neural stem cells. Cell Stem Cell 2012; 10: 473-9.
[25]
Nagoshi N, Okano H. iPSC-derived neural precursor cells: Potential for cell transplantation therapy in spinal cord injury. Cell Mol Life Sci 2018; 75: 989-1000.
[26]
Reynolds B, Weiss S. Generation of neurons and astrocytes from isolated cells of the adult mammalian central nervous system. Science 1992; 255: 1707-10.
[27]
Karow M, Camp J, Falk S, et al. Direct pericyte-to-neuron reprogramming via unfolding of a neural stem cell-like program. Nat Neurosci 2018; 21: 932-40.
[28]
Cairns D, Chwalek K, Moore Y, et al. Expandable and rapidly differentiating human induced neural stem cell lines for multiple tissue engineering applications. Stem Cell Reports 2016; 7: 557-70.
[29]
Takahashi K, Tanabe K, Ohnuki M, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 2007; 131: 861-72.
[30]
Zhang S, Wernig M, Duncan I, Brüstle O, Thomson J. In vitro differentiation of transplantable neural precursors from human embryonic stem cells. Nat Biotechnol 2001; 19: 1129-33.
[31]
Dhara S, Stice S. Neural differentiation of human embryonic stem cells. J Cell Biochem 2008; 105: 633-40.
[32]
Yuan S, Martin J, Elia J, et al. Cell-surface marker signatures for the isolation of neural stem cells, glia and neurons derived from human pluripotent stem cells. PLoS ONE 2011; 6: e17540.
[33]
Hofrichter M, Nimtz L, Tigges J, et al. Comparative performance analysis of human iPSC-derived and primary neural progenitor cells (NPC) grown as neurospheres in vitro. Stem Cell Res 2017; 25: 72-82.
[34]
Kang S, Chen X, Gong S, et al. Characteristic analyses of a neural differentiation model from iPSC-derived neuron according to morphology, physiology, and global gene expression pattern. Scientific reports 2017; 7: 12233.
[35]
Okubo T, Nagoshi N, Kohyama J, et al. Treatment with a gamma-secretase inhibitor promotes functional recovery in human iPSC- derived transplants for chronic spinal cord injury. Stem Cell Reports 2018; 11: 1416-32.
[36]
Chambers S, Fasano C, Papapetrou E, Tomishima M, Sadelain M, Studer L. Highly efficient neural conversion of human ES and iPS cells by dual inhibition of SMAD signaling. Nat Biotechnol 2009; 27: 275-80.
[37]
Yang D, Li T, Xu M, et al. Graphene oxide promotes the differentiation of mouse embryonic stem cells to dopamine neurons. Nanomedicine 2014; 9: 2445-55.
[38]
Choi HW, Hong YJ, Kim JS, et al. In vivo differentiation of induced pluripotent stem cells into neural stem cells by chimera formation. PLoS One 2017; 12: e0170735.
[39]
Numasawa-Kuroiwa Y, Okada Y, Shibata S, et al. Involvement of ER stress in dysmyelination of Pelizaeus-Merzbacher Disease with PLP1 missense mutations shown by iPSC-derived oligodendrocytes. Stem Cell Reports 2014; 2: 648-61.
[40]
Kumamaru H, Kadoya K, Adler A, et al. Generation and post-injury integration of human spinal cord neural stem cells. Nat Methods 2018; 15(9): 723-31.
[41]
Bain G, Kitchens D, Yao M, Huettner J, Gottlieb D. Embryonic stem cells express neuronal properties in vitro. Dev Biol 1995; 168: 342-57.
[42]
Carson C, Aigner S, Gage F. Stem cells: The good, bad and barely in control. Nat Med 2006; 12: 1237-8.
[43]
Yeo G, Xu X, Liang T, et al. Alternative splicing events identified in human embryonic stem cells and neural progenitors. PLOS Comput Biol 2007; 3: 1951-67.
[44]
Reinhardt P, Glatza M, Hemmer K, et al. Derivation and expansion using only small molecules of human neural progenitors for neurodegenerative disease modeling. PLoS One 2013; 8: e59252.
[45]
Neely M, Litt M, Tidball A, et al. DMH1, a highly selective small molecule BMP inhibitor promotes neurogenesis of hiPSCs: Comparison of PAX6 and SOX1 expression during neural induction. ACS Chem Neurosci 2012; 3: 482-91.
[46]
Fünfschilling U, Supplie L, Mahad D, et al. Glycolytic oligodendrocytes maintain myelin and long-term axonal integrity. Nature 2012; 485: 517-21.
[47]
Khazaei M, Ahuja C, Fehlings M. Generation of oligodendrogenic spinal neural progenitor cells from human induced pluripotent stem Cells. Curr Protoc Stem Cell BiolM 2017; 42: 2D.20.1-2D. 14.
[48]
D’Aiuto L, Zhi Y, Kumar Das D, et al. Large-scale generation of human iPSC-derived neural stem cells/early neural progenitor cells and their neuronal differentiation. Organogenesis 2014; 10: 365-77.
[49]
Li X, Du Z, Zarnowska E, et al. Specification of motoneurons from human embryonic stem cells. Nat Biotechnol 2005; 23: 215-21.
[50]
Chandrasekaran A, Avci H, Ochalek A, et al. Comparison of 2D and 3D neural induction methods for the generation of neural progenitor cells from human induced pluripotent stem cells. Stem Cell Res 2017; 25: 139-51.
[51]
Perrier AL, Tabar V, Barberi T, et al. Derivation of midbrain dopamine neurons from human embryonic stem cells. Proc Natl Acad Sci USA 2004; 101: 12543-8.
[52]
Assinck P, Duncan GJ, Hilton BJ, Plemel JR, Tetzlaff W. Cell transplantation therapy for spinal cord injury. Nat Neurosci 2017; 20: 637-47.
[53]
Nagoshi N, Okano H. Applications of induced pluripotent stem cell technologies in spinal cord injury. J Neurochem 2017; 141: 848-60.
[54]
Salewski R, Buttigieg J, Mitchell R, van der Kooy D, Nagy A, Fehlings M. The generation of definitive neural stem cells from PiggyBac transposon-induced pluripotent stem cells can be enhanced by induction of the NOTCH signaling pathway. Stem Cells Dev 2013; 22: 383-96.
[55]
Kumagai G, Okada Y, Yamane J, et al. Roles of ES cell-derived gliogenic neural stem/progenitor cells in functional recovery after spinal cord injury. PLoS ONE 2009; 4: e7706.
[56]
Yousefifard M, Rahimi-Movaghar V, Nasirinezhad F, et al. Neural stem/progenitor cell transplantation for spinal cord injury treatment; A systematic review and meta-analysis. Neuroscience 2016; 322: 377-97.
[57]
Lee YS, Lin CY, Jiang HH, Depaul M, Lin VW, Silver J. Nerve regeneration restores supraspinal control of bladder function after complete spinal cord injury. J Neurosci 2013; 33: 10591-606.
[58]
Tuszynski MH, Steward O. Concepts and methods for the study of axonal regeneration in the CNS. Neuron 2012; 74: 777-91.
[59]
Liu K, Lu Y, Lee J, et al. PTEN deletion enhances the regenerative ability of adult corticospinal neurons. Nat Neurosci 2010; 13: 1075-81.
[60]
Starkey M, Schwab M. Anti-Nogo-A and training: Can one plus one equal three? Exp Neurol 2012; 235: 53-61.
[61]
Nutt S, Chang E, Suhr S, et al. Caudalized human iPSC-derived neural progenitor cells produce neurons and glia but fail to restore function in an early chronic spinal cord injury model. Exp Neurol 2013; 248: 491-503.
[62]
Verrier L, Davidson L, Gierlinski M, Dady A, Storey KG. Neural differentiation, selection and transcriptomic profiling of human neuromesodermal progenitor-like cells in vitro. Development 2018; 145: dev166215.
[63]
Tzouanacou E, Wegener A, Wymeersch FJ, Wilson V, Nicolas JF. Redefining the progression of lineage segregations during mammalian embryogenesis by clonal analysis. Dev Cell 2009; 17: 365-76.
[64]
Ogura T, Sakaguchi H, Miyamoto S, Takahashi J. Three-dimensional induction of dorsal, intermediate and ventral spinal cord tissues from human pluripotent stem cells. Development 2018; 145: dev162214.
[65]
Levi AD, Okonkwo DO, Park P, et al. Emerging safety of intramedullary transplantation of human neural stem cells in chronic cervical and thoracic spinal cord injury. Neurosurgery 2018; 82: 562-75.
[66]
Levi AD, Anderson KD, Okonkwo DO, et al. Clinical outcomes from a multi-center study of human neural stem cell transplantation in chronic cervical spinal cord Injury. J Neurotrauma 2019; 36(6): 891-902.
[67]
Curtis E, Martin JR, Gabel B, et al. A first-in-human, phase i study of neural stem cell transplantation for chronic spinal cord Injury. Cell Stem Cell 2018; 22: 941-50.e6.
[68]
Zhang M, Ngo J, Pirozzi F, Sun Y, Wynshaw-Boris A. Highly efficient methods to obtain homogeneous dorsal neural progenitor cells from human and mouse embryonic stem cells and induced pluripotent stem cells. Stem Cell Res Ther 2018; 9(1): 67.
[69]
Shi Y, Kirwan P, Livesey F. Directed differentiation of human pluripotent stem cells to cerebral cortex neurons and neural networks. Nat Protoc 2012; 7: 1836-46.
[70]
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.
[71]
Bibel M, Richter J, Schrenk K, et al. Differentiation of mouse embryonic stem cells into a defined neuronal lineage. Nat Neurosci 2004; 7: 1003-9.

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy