Neonatal Med.  2019 Feb;26(1):1-16. 10.5385/nm.2019.26.1.1.

Human Neural Stem Cells: Translational Research for Neonatal Hypoxic-Ischemic Brain Injury

Affiliations
  • 1Division of Neonatology, Department of Pediatrics, Severance Children's Hospital, Yonsei University College of Medicine, Seoul, Korea. kipark@yuhs.ac
  • 2BK21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea.

Abstract

Neonatal hypoxic-ischemic (HI) brain injury is a major cause of neonatal mortality and long-term neurodevelopmental disabilities. Although promising neuroprotective interventions have been studied, the current management of HI brain injury has been limited to supportive measures and induced hypothermia. In addition to engrafting, migrating toward the damage sites and differentiating into multiple lineages, multipotent neural stem/progenitor cells (NSPCs) also provide trophic/immunomodulatory factors and integrate into the host neurons upon implantation into an HI-injured brain. However, NSPC-based therapies have shown poor cell survival and integration, poor differentiation or restricted differentiation into the glial lineages. Furthermore, to achieve full functional recovery following brain injury, the optimization of cell therapy is needed to recapitulate the precise migration of stem cells to the region of interest and the neural rewiring present in the brain microenvironment. Therefore, the efficacy of NSPCs in the treatment of CNS injury is currently insufficient. Human NSPCs (hNSPCs) were isolated from the forebrain of an aborted fetus at 13 weeks of gestation with full parental consent and the approval of the Institutional Review Board of the Yonsei University College of Medicine. Here, to enhance the regenerative ability of hNSPCs in HI brain injury, cells were either pretreated with pharmacological agents or engineered to serve as vehicles for gene delivery. Furthermore, when combined with a poly (glycolic acid)-based synthetic scaffold, hNSPCs provide a more versatile treatment for neonatal HI brain injury. Finally, hNSPCs transfected with zinc-doped ferrite magnetic nanoparticles for controlling both cell migration and differentiation offer a simple and smart tool for cell-based therapies.

Keyword

Neural stem cells; Hypoxia-ischemia, brain; Cell therapy; Genetic therapy

MeSH Terms

Aborted Fetus
Brain Injuries*
Brain*
Cell Movement
Cell Survival
Cell- and Tissue-Based Therapy
Ethics Committees, Research
Genetic Therapy
Humans*
Hypothermia, Induced
Hypoxia-Ischemia, Brain
Infant
Infant Mortality
Nanoparticles
Neural Stem Cells*
Neurons
Parental Consent
Pregnancy
Prosencephalon
Stem Cells
Translational Medical Research*

Figure

  • Figure 1. Transplantation (Tx) of human nucleus pulposus cell (hNPC)-poly (glycolic acid) (PGA) complexes into the infarction cavity of a hypoxic-ischemic (HI) brain injury. (A) Two weeks following Tx of the hNPC-PGA complexes (an arrow) into the infarction cavity indicated by an asterisk; human nuclei (hNuc)+donor-derived cells (green) show robust engraftment within the injured area. The PGA fibers begin to biodegrade (green strands, arrowheads). (B-G) Some human neural stem/progenitor cell (hNSPC)+donor-derived cells (red in B, D, E, G) presented co-localization with neurofilament (NF) (blue arrows in B, C, D) with either extended neuronal processes (white arrows in C, D; n=15) or glial fibrillary acidic protein (GFAP) (white arrows in E, F, G; n=15). The PGA fibers are indicated by arrowheads in B-G. The tract tracer biotinylated dextran amine conjugated with fluorescein (BDA-FITC) was injected into the contralateral intact cortex at 10 weeks following implantation of the hNPC-PGA complex into the infarction cavity. Axonal projections were labeled in green with fluorescein and can be visualized from the cerebral cortex as well as the external capsule (EC) of the intact hemisphere, projecting through the corpus callosum (CC) and toward the implantation site of the hNPC-PGA complex within the injured ipsilateral cortex and penumbra (H). Some BDA-FITC+anterograde-labeled processes from the neurons in the contralateral intact hemisphere were observed to sprout their axons toward the implantation site of the hNPC-PGA complex (H). The BDA-FITC+retrograde-labeled cell body and cellular processes of a neuron-like cell in the ipsilateral cortical penumbra indicated by an asterisk (in H) are well visualized at a higher magnification (I). The data shown are representative images. Scale bars (A, H: 500 μm; B, E: 20 μm).


Reference

1. Ferriero DM. Neonatal brain injury. N Engl J Med. 2004; 351:1985–95.
2. Volpe JJ. Neonatal encephalopathy: an inadequate term for hypoxic- ischemic encephalopathy. Ann Neurol. 2012; 72:156–66.
3. Douglas-Escobar M, Weiss MD. Hypoxic-ischemic encephalopathy: a review for the clinician. JAMA Pediatr. 2015; 169:397–403.
4. Conti L, Cattaneo E. Neural stem cell systems: physiological players or in vitro entities? Nat Rev Neurosci. 2010; 11:176–87.
5. McKay R. Stem cells in the central nervous system. Science. 1997; 276:66–71.
6. Gage FH. Mammalian neural stem cells. Science. 2000; 287:1433–8.
7. Ourednik V, Ourednik J, Flax JD, Zawada WM, Hutt C, Yang C, et al. Segregation of human neural stem cells in the developing primate forebrain. Science. 2001; 293:1820–4.
8. Svendsen CN, ter Borg MG, Armstrong RJ, Rosser AE, Chandran S, Ostenfeld T, et al. A new method for the rapid and long term growth of human neural precursor cells. J Neurosci Methods. 1998; 85:141–52.
9. Vescovi AL, Parati EA, Gritti A, Poulin P, Ferrario M, Wanke E, et al. Isolation and cloning of multipotential stem cells from the embryonic human CNS and establishment of transplantable human neural stem cell lines by epigenetic stimulation. Exp Neurol. 1999; 156:71–83.
10. Palmer TD, Ray J, Gage FH. FGF-2-responsive neuronal progenitors reside in proliferative and quiescent regions of the adult rodent brain. Mol Cell Neurosci. 1995; 6:474–86.
11. Reynolds BA, Weiss S. Generation of neurons and astrocytes from isolated cells of the adult mammalian central nervous system. Science. 1992; 255:1707–10.
12. Martino G, Pluchino S. The therapeutic potential of neural stem cells. Nat Rev Neurosci. 2006; 7:395–406.
13. Abematsu M, Tsujimura K, Yamano M, Saito M, Kohno K, Kohyama J, et al. Neurons derived from transplanted neural stem cells restore disrupted neuronal circuitry in a mouse model of spinal cord injury. J Clin Invest. 2010; 120:3255–66.
14. Kelly S, Bliss TM, Shah AK, Sun GH, Ma M, Foo WC, et al. Transplanted human fetal neural stem cells survive, migrate, and differentiate in ischemic rat cerebral cortex. Proc Natl Acad Sci U S A. 2004; 101:11839–44.
15. Park KI, Teng YD, Snyder EY. The injured brain interacts reciprocally with neural stem cells supported by scaffolds to reconstitute lost tissue. Nat Biotechnol. 2002; 20:1111–7.
16. Blurton-Jones M, Kitazawa M, Martinez-Coria H, Castello NA, Muller FJ, Loring JF, et al. Neural stem cells improve cognition via BDNF in a transgenic model of Alzheimer disease. Proc Natl Acad Sci U S A. 2009; 106:13594–9.
17. Pluchino S, Zanotti L, Rossi B, Brambilla E, Ottoboni L, Salani G, et al. Neurosphere-derived multipotent precursors promote neuroprotection by an immunomodulatory mechanism. Nature. 2005; 436:266–71.
18. Tamaki SJ, Jacobs Y, Dohse M, Capela A, Cooper JD, Reitsma M, et al. Neuroprotection of host cells by human central nervous system stem cells in a mouse model of infantile neuronal ceroid lipofuscinosis. Cell Stem Cell. 2009; 5:310–9.
19. Andres RH, Horie N, Slikker W, Keren-Gill H, Zhan K, Sun G, et al. Human neural stem cells enhance structural plasticity and axonal transport in the ischaemic brain. Brain. 2011; 134:1777–89.
20. Gage FH, Temple S. Neural stem cells: generating and regenerating the brain. Neuron. 2013; 80:588–601.
21. Daadi MM, Davis AS, Arac A, Li Z, Maag AL, Bhatnagar R, et al. Human neural stem cell grafts modify microglial response and enhance axonal sprouting in neonatal hypoxic-ischemic brain injury. Stroke. 2010; 41:516–23.
22. Imitola J, Raddassi K, Park KI, Mueller FJ, Nieto M, Teng YD, et al. Directed migration of neural stem cells to sites of CNS injury by the stromal cell-derived factor 1alpha/CXC chemokine receptor 4 pathway. Proc Natl Acad Sci U S A. 2004; 101:18117–22.
23. Park KI, Hack MA, Ourednik J, Yandava B, Flax JD, Stieg PE, et al. Acute injury directs the migration, proliferation, and differentiation of solid organ stem cells: evidence from the effect of hypoxia-ischemia in the CNS on clonal "reporter" neural stem cells. Exp Neurol. 2006; 199:156–78.
24. Park KI, Himes BT, Stieg PE, Tessler A, Fischer I, Snyder EY. Neural stem cells may be uniquely suited for combined gene therapy and cell replacement: evidence from engraftment of Neurotrophin-3-expressing stem cells in hypoxic-ischemic brain injury. Exp Neurol. 2006; 199:179–90.
25. Lindvall O, Kokaia Z. Stem cells in human neurodegenerative disorders: time for clinical translation? J Clin Invest. 2010; 120:29–40.
26. Barnabe-Heider F, Frisen J. Stem cells for spinal cord repair. Cell Stem Cell. 2008; 3:16–24.
27. Orive G, Anitua E, Pedraz JL, Emerich DF. Biomaterials for promoting brain protection, repair and regeneration. Nat Rev Neurosci. 2009; 10:682–92.
28. Hofstetter CP, Holmstrom NA, Lilja JA, Schweinhardt P, Hao J, Spenger C, et al. Allodynia limits the usefulness of intraspinal neural stem cell grafts: directed differentiation improves outcome. Nat Neurosci. 2005; 8:346–53.
29. Hicks AU, Lappalainen RS, Narkilahti S, Suuronen R, Corbett D, Sivenius J, et al. Transplantation of human embryonic stem cellderived neural precursor cells and enriched environment after cortical stroke in rats: cell survival and functional recovery. Eur J Neurosci. 2009; 29:562–74.
30. Nakagomi N, Nakagomi T, Kubo S, Nakano-Doi A, Saino O, Takata M, et al. Endothelial cells support survival, proliferation, and neuronal differentiation of transplanted adult ischemiainduced neural stem/progenitor cells after cerebral infarction. Stem Cells. 2009; 27:2185–95.
31. Park KI, Goo K, Jung K, Kim M, Kim IS, Yun S, et al. Therapeutic application of neural stem cells for neonatal hypoxic-ischemic brain injury. Neonatal Med. 2013; 20:343–53.
32. Wilkinson G, Dennis D, Schuurmans C. Proneural genes in neocortical development. Neuroscience. 2013; 253:256–73.
33. Serre A, Snyder EY, Mallet J, Buchet D. Overexpression of basic helix-loop-helix transcription factors enhances neuronal differentiation of fetal human neural progenitor cells in various ways. Stem Cells Dev. 2012; 21:539–53.
34. Yi SH, Jo AY, Park CH, Koh HC, Park RH, Suh-Kim H, et al. Mash1 and neurogenin 2 enhance survival and differentiation of neural precursor cells after transplantation to rat brains via distinct modes of action. Mol Ther. 2008; 16:1873–82.
35. Lee IS, Koo KY, Jung K, Kim M, Kim IS, Hwang K, et al. Neurogenin-2-transduced human neural progenitor cells attenuate neonatal hypoxic-ischemic brain injury. Transl Res. 2017; 183:121–36.
36. Hagberg H, Mallard C, Ferriero DM, Vannucci SJ, Levison SW, Vexler ZS, et al. The role of inflammation in perinatal brain injury. Nat Rev Neurol. 2015; 11:192–208.
37. Banjara M, Ghosh C. Sterile neuro inflammation and strategies for therapeutic intervention. Int J Inflam. 2017; 2017:8385961.
38. Pluchino S, Quattrini A, Brambilla E, Gritti A, Salani G, Dina G, et al. Injection of adult neurospheres induces recovery in a chronic model of multiple sclerosis. Nature. 2003; 422:688–94.
39. Einstein O, Fainstein N, Vaknin I, Mizrachi-Kol R, Reihartz E, Grigoriadis N, et al. Neural precursors attenuate autoimmune encephalomyelitis by peripheral immunosuppression. Ann Neurol. 2007; 61:209–18.
40. Lee IS, Jung K, Kim IS, Lee H, Kim M, Yun S, et al. Human neural stem cells alleviate Alzheimer-like pathology in a mouse model. Mol Neurodegener. 2015; 10:38.
41. Kwilasz AJ, Grace PM, Serbedzija P, Maier SF, Watkins LR. The therapeutic potential of interleukin-10 in neuroimmune diseases. Neuropharmacology. 2015; 96:55–69.
42. de Bilbao F, Arsenijevic D, Moll T, Garcia-Gabay I, Vallet P, Langhans W, et al. In vivo over-expression of interleukin-10 increases resistance to focal brain ischemia in mice. J Neurochem. 2009; 110:12–22.
43. Ooboshi H, Ibayashi S, Shichita T, Kumai Y, Takada J, Ago T, et al. Postischemic gene transfer of interleukin-10 protects against both focal and global brain ischemia. Circulation. 2005; 111:913–9.
44. Grilli M, Barbieri I, Basudev H, Brusa R, Casati C, Lozza G, et al. Interleukin-10 modulates neuronal threshold of vulnerability to ischaemic damage. Eur J Neurosci. 2000; 12:2265–72.
45. Vila N, Castillo J, Davalos A, Esteve A, Planas AM, Chamorro A. Levels of anti-inflammatory cytokines and neurological worsening in acute ischemic stroke. Stroke. 2003; 34:671–5.
46. Kettenmann H, Hanisch UK, Noda M, Verkhratsky A. Physiology of microglia. Physiol Rev. 2011; 91:461–553.
47. Block ML, Zecca L, Hong JS. Microglia-mediated neurotoxicity: uncovering the molecular mechanisms. Nat Rev Neurosci. 2007; 8:57–69.
48. Dheen ST, Kaur C, Ling EA. Microglial activation and its implications in the brain diseases. Curr Med Chem. 2007; 14:1189–97.
49. Iadecola C, Anrather J. The immunology of stroke: from mechanisms to translation. Nat Med. 2011; 17:796–808.
50. Hanisch UK, Kettenmann H. Microglia: active sensor and versatile effector cells in the normal and pathologic brain. Nat Neurosci. 2007; 10:1387–94.
51. Kwon MJ, Kim J, Shin H, Jeong SR, Kang YM, Choi JY, et al. Contribution of macrophages to enhanced regenerative capacity of dorsal root ganglia sensory neurons by conditioning injury. J Neurosci. 2013; 33:15095–108.
52. Miron VE, Boyd A, Zhao JW, Yuen TJ, Ruckh JM, Shadrach JL, et al. M2 microglia and macrophages drive oligodendrocyte differentiation during CNS remyelination. Nat Neurosci. 2013; 16:1211–8.
53. Thored P, Heldmann U, Gomes-Leal W, Gisler R, Darsalia V, Taneera J, et al. Long-term accumulation of microglia with proneurogenic phenotype concomitant with persistent neurogenesis in adult subventricular zone after stroke. Glia. 2009; 57:835–49.
54. Ekdahl CT, Claasen JH, Bonde S, Kokaia Z, Lindvall O. Inflammation is detrimental for neurogenesis in adult brain. Proc Natl Acad Sci U S A. 2003; 100:13632–7.
55. Liu Z, Fan Y, Won SJ, Neumann M, Hu D, Zhou L, et al. Chronic treatment with minocycline preserves adult new neurons and reduces functional impairment after focal cerebral ischemia. Stroke. 2007; 38:146–52.
56. David S, Kroner A. Repertoire of microglial and macrophage responses after spinal cord injury. Nat Rev Neurosci. 2011; 12:388–99.
57. Mosser DM, Edwards JP. Exploring the full spectrum of macrophage activation. Nat Rev Immunol. 2008; 8:958–69.
58. Rosenblum S, Smith TN, Wang N, Chua JY, Westbroek E, Wang K, et al. BDNF pretreatment of human embryonic-derived neural stem cells improves cell survival and functional recovery after transplantation in hypoxic-ischemic stroke. Cell Transplant. 2015; 24:2449–61.
59. Hallenbeck JM. The many faces of tumor necrosis factor in stroke. Nat Med. 2002; 8:1363–8.
60. Liu H, Honmou O, Harada K, Nakamura K, Houkin K, Hamada H, et al. Neuroprotection by PlGF gene-modified human mesenchymal stem cells after cerebral ischaemia. Brain. 2006; 129:2734–45.
61. Lee HJ, Kim MK, Kim HJ, Kim SU. Human neural stem cells genetically modified to overexpress Akt1 provide neuroprotection and functional improvement in mouse stroke model. PLoS One. 2009; 4:e5586.
62. Sakata H, Niizuma K, Wakai T, Narasimhan P, Maier CM, Chan PH. Neural stem cells genetically modified to overexpress cu/zn-superoxide dismutase enhance amelioration of ischemic stroke in mice. Stroke. 2012; 43:2423–9.
63. Wang F, Kameda M, Yasuhara T, Tajiri N, Kikuchi Y, Liang HB, et al. GDNF-pretreatment enhances the survival of neural stem cells following transplantation in a rat model of Parkinson's disease. Neurosci Res. 2011; 71:92–8.
64. Sakata H, Narasimhan P, Niizuma K, Maier CM, Wakai T, Chan PH. Interleukin 6-preconditioned neural stem cells reduce ischaemic injury in stroke mice. Brain. 2012; 135:3298–310.
65. Schwabe RF, Brenner DA. Mechanisms of liver injury. I. TNFalpha-induced liver injury: role of IKK, JNK, and ROS pathways. Am J Physiol Gastrointest Liver Physiol. 2006; 290:G583–9.
66. Turrin NP, Rivest S. Tumor necrosis factor alpha but not interleukin 1 beta mediates neuroprotection in response to acute nitric oxide excitotoxicity. J Neurosci. 2006; 26:143–51.
67. Bernardino L, Agasse F, Silva B, Ferreira R, Grade S, Malva JO. Tumor necrosis factor-alpha modulates survival, proliferation, and neuronal differentiation in neonatal subventricular zone cell cultures. Stem Cells. 2008; 26:2361–71.
68. Peng H, Whitney N, Wu Y, Tian C, Dou H, Zhou Y, et al. HIV-1-infected and/or immune-activated macrophage-secreted TNFalpha affects human fetal cortical neural progenitor cell proliferation and differentiation. Glia. 2008; 56:903–16.
69. Lan X, Chen Q, Wang Y, Jia B, Sun L, Zheng J, et al. TNF-α affects human cortical neural progenitor cell differentiation through the autocrine secretion of leukemia inhibitory factor. PLoS One. 2012; 7:e50783.
70. Cuschieri J, Maier RV. Mitogen-activated protein kinase (MAPK). Crit Care Med. 2005; 33:S417–9.
71. Kim M, Jung K, Kim IS, Lee IS, Ko Y, Shin JE, et al. TNF-α induces human neural progenitor cell survival after oxygen-glucose deprivation by activating the NF-κB pathway. Exp Mol Med. 2018; 50:14.
72. Garcia GE, Xia Y, Chen S, Wang Y, Ye RD, Harrison JK, et al. NFkappaB-dependent fractalkine induction in rat aortic endothelial cells stimulated by IL-1beta, TNF-alpha, and LPS. J Leukoc Biol. 2000; 67:577–84.
73. Fraticelli P, Sironi M, Bianchi G, D'Ambrosio D, Albanesi C, Stoppacciaro A, et al. Fractalkine (CX3CL1) as an amplification circuit of polarized Th1 responses. J Clin Invest. 2001; 107:1173–81.
74. Zujovic V, Benavides J, Vige X, Carter C, Taupin V. Fractalkine modulates TNF-alpha secretion and neurotoxicity induced by microglial activation. Glia. 2000; 29:305–15.
75. Cardona AE, Pioro EP, Sasse ME, Kostenko V, Cardona SM, Dijkstra IM, et al. Control of microglial neurotoxicity by the fractalkine receptor. Nat Neurosci. 2006; 9:917–24.
76. Teng YD, Lavik EB, Qu X, Park KI, Ourednik J, Zurakowski D, et al. Functional recovery following traumatic spinal cord injury mediated by a unique polymer scaffold seeded with neural stem cells. Proc Natl Acad Sci U S A. 2002; 99:3024–9.
77. Lee S, Yun S, Park KI, Jang JH. Sliding fibers: slidable, injectable, and gel-like electrospun nanofibers as versatile cell carriers. ACS Nano. 2016; 10:3282–94.
78. Skop NB, Calderon F, Cho CH, Gandhi CD, Levison SW. Improvements in biomaterial matrices for neural precursor cell transplantation. Mol Cell Ther. 2014; 2:19.
79. Silver J, Miller JH. Regeneration beyond the glial scar. Nat Rev Neurosci. 2004; 5:146–56.
80. Fitch MT, Doller C, Combs CK, Landreth GE, Silver J. Cellular and molecular mechanisms of glial scarring and progressive cavitation: in vivo and in vitro analysis of inflammation-induced secondary injury after CNS trauma. J Neurosci. 1999; 19:8182–98.
81. Stone BS, Zhang J, Mack DW, Mori S, Martin LJ, Northington FJ. Delayed neural network degeneration after neonatal hypoxiaischemia. Ann Neurol. 2008; 64:535–46.
82. Barrett RD, Bennet L, Davidson J, Dean JM, George S, Emerald BS, et al. Destruction and reconstruction: hypoxia and the developing brain. Birth Defects Res C Embryo Today. 2007; 81:163–76.
83. Huang YC, Huang YY. Biomaterials and strategies for nerve regeneration. Artif Organs. 2006; 30:514–22.
84. Lu P, Wang Y, Graham L, McHale K, Gao M, Wu D, et al. Longdistance growth and connectivity of neural stem cells after severe spinal cord injury. Cell. 2012; 150:1264–73.
85. Zeng X, Zeng YS, Ma YH, Lu LY, Du BL, Zhang W, et al. Bone marrow mesenchymal stem cells in a three-dimensional gelatin sponge scaffold attenuate inflammation, promote angiogenesis, and reduce cavity formation in experimental spinal cord injury. Cell Transplant. 2011; 20:1881–99.
86. Olson HE, Rooney GE, Gross L, Nesbitt JJ, Galvin KE, Knight A, et al. Neural stem cell- and Schwann cell-loaded biodegradable polymer scaffolds support axonal regeneration in the transected spinal cord. Tissue Eng Part A. 2009; 15:1797–805.
87. Cholas RH, Hsu HP, Spector M. The reparative response to crosslinked collagen-based scaffolds in a rat spinal cord gap model. Biomaterials. 2012; 33:2050–9.
88. Hejcl A, Sedy J, Kapcalova M, Toro DA, Amemori T, Lesy P, et al. HPMA-RGD hydrogels seeded with mesenchymal stem cells improve functional outcome in chronic spinal cord injury. Stem Cells Dev. 2010; 19:1535–46.
89. Lee KY, Kong HJ, Mooney DJ. Quantifying interactions between cell receptors and adhesion ligand-modified polymers in solution. Macromol Biosci. 2008; 8:140–5.
90. Karp JM, Leng Teo GS. Mesenchymal stem cell homing: the devil is in the details. Cell Stem Cell. 2009; 4:206–16.
91. Cheng Z, Ou L, Zhou X, Li F, Jia X, Zhang Y, et al. Targeted migration of mesenchymal stem cells modified with CXCR4 gene to infarcted myocardium improves cardiac performance. Mol Ther. 2008; 16:571–9.
92. Rosova I, Dao M, Capoccia B, Link D, Nolta JA. Hypoxic preconditioning results in increased motility and improved therapeutic potential of human mesenchymal stem cells. Stem Cells. 2008; 26:2173–82.
93. Barbash IM, Chouraqui P, Baron J, Feinberg MS, Etzion S, Tessone A, et al. Systemic delivery of bone marrow-derived mesenchymal stem cells to the infarcted myocardium: feasibility, cell migration, and body distribution. Circulation. 2003; 108:863–8.
94. Purcell BP, Elser JA, Mu A, Margulies KB, Burdick JA. Synergistic effects of SDF-1α chemokine and hyaluronic acid release from degradable hydrogels on directing bone marrow derived cell homing to the myocardium. Biomaterials. 2012; 33:7849–57.
95. Park JS, Rhau B, Hermann A, McNally KA, Zhou C, Gong D, et al. Synthetic control of mammalian-cell motility by engineering chemotaxis to an orthogonal bioinert chemical signal. Proc Natl Acad Sci U S A. 2014; 111:5896–901.
96. Shin TH, Cheon J. Synergism of nanomaterials with physical stimuli for biology and medicine. Acc Chem Res. 2017; 50:567–72.
97. Riegler J, Liew A, Hynes SO, Ortega D, O'Brien T, Day RM, et al. Superparamagnetic iron oxide nanoparticle targeting of MSCs in vascular injury. Biomaterials. 2013; 34:1987–94.
98. Landazuri N, Tong S, Suo J, Joseph G, Weiss D, Sutcliffe DJ, et al. Magnetic targeting of human mesenchymal stem cells with internalized superparamagnetic iron oxide nanoparticles. Small. 2013; 9:4017–26.
99. Vandergriff AC, Hensley TM, Henry ET, Shen D, Anthony S, Zhang J, et al. Magnetic targeting of cardiosphere-derived stem cells with ferumoxytol nanoparticles for treating rats with myocardial infarction. Biomaterials. 2014; 35:8528–39.
100. Tukmachev D, Lunov O, Zablotskii V, Dejneka A, Babic M, Sykova E, et al. An effective strategy of magnetic stem cell delivery for spinal cord injury therapy. Nanoscale. 2015; 7:3954–8.
101. Yun S, Shin TH, Lee JH, Cho MH, Kim IS, Kim JW, et al. Design of magnetically labeled cells (mag-cells) for in vivo control of stem cell migration and differentiation. Nano Lett. 2018; 18:838–45.
102. Lledo PM, Alonso M, Grubb MS. Adult neurogenesis and functional plasticity in neuronal circuits. Nat Rev Neurosci. 2006; 7:179–93.
103. Sandoval KE, Witt KA. Blood-brain barrier tight junction permeability and ischemic stroke. Neurobiol Dis. 2008; 32:200–19.
Full Text Links
  • NM
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr