441
Views
14
CrossRef citations to date
0
Altmetric
Review

Advances in stem cell treatment for sciatic nerve injury

ORCID Icon, ORCID Icon & ORCID Icon
Pages 301-311 | Received 17 Sep 2018, Accepted 28 Jan 2019, Published online: 11 Feb 2019

References

  • Chen Z-L, Yu W-M, Strickland S. Peripheral regeneration. Annu Rev Neurosci. 2007;30:209–233.
  • Lundborg G. The nerve trunk. nerve injury and repair. 2nd ed. London: Churchill Livingstone; 1988. p. 32–63.
  • Griffin JW, Hogan MV, Chhabra AB, et al. Peripheral nerve repair and reconstruction. Jbjs. 2013;95(23):2144–2151.
  • Trojaborg W. Rate of recovery in motor and sensory fibers of the radial nerve: clinical and electrophysiological aspects. J Neurol Neurosurg. 1970;33(5):625–638.
  • Burnett MG, Zager EL. Pathophysiology of peripheral nerve injury: a brief review. Neurosurg Focus. 2004;16(5):1–7.
  • Grinsell D, Keating CP. Peripheral nerve reconstruction after injury: a review of clinical and experimental therapies. Biomed Res Int. 2014;2014:698256.
  • Ryu J, Beimesch CF, Lalli TJ. (iii) Peripheral nerve repair. Orthop Trauma. 2011;25(3):174–180.
  • Pfister BJ, Gordon T, Loverde JR, et al. Biomedical engineering strategies for peripheral nerve repair: surgical applications, state of the art, and future challenges. Crit Rev Biomed Eng. 2011;39:2.
  • Siemionow M, Brzezicki G. Current techniques and concepts in peripheral nerve repair. Int Rev Neurobiol. 2009;87:141–172.
  • Zhang Y, Luo H, Zhang Z, et al. A nerve graft constructed with xenogeneic acellular nerve matrix and autologous adipose-derived mesenchymal stem cells. Biomaterials. 2010;31(20):5312–5324.
  • Thomson JA, Itskovitz-Eldor J, Shapiro SS, et al. Embryonic stem cell lines derived from human blastocysts. Science (New York, NY). 1998;282(5391):1145–1147.
  • Lee G, Chambers SM, Tomishima MJ, et al. Derivation of neural crest cells from human pluripotent stem cells. Nat Protoc. 2010;5(4):688.
  • Cui L, Jiang J, Wei L, et al. Transplantation of embryonic stem cells improves nerve repair and functional recovery after severe sciatic nerve axotomy in rats. Stem Cells. 2008;26(5):1356–1365.
  • Mozafari R, Kyrylenko S, Castro MV, et al. Combination of heterologous fibrin sealant and bioengineered human embryonic stem cells to improve regeneration following autogenous sciatic nerve grafting repair. J Venomous Anim Toxins incl Trop Dis. 2018;24(1):11.
  • Jones I, Novikova LN, Novikov LN, et al. Regenerative effects of human embryonic stem cell‐derived neural crest cells for treatment of peripheral nerve injury. J Tissue Eng Regen Med. 2018;12(4):e2099–e109.
  • Li Y, Guo L, Ahn HS, et al. Amniotic mesenchymal stem cells display neurovascular tropism and aid in the recovery of injured peripheral nerves. J Cell Mol Med. 2014;18(6):1028–1034.
  • Cheng F-C, Tai M-H, Sheu M-L, et al. Enhancement of regeneration with glia cell line-derived neurotrophic factor–transduced human amniotic fluid mesenchymal stem cells after sciatic nerve crush injury [RETRACTED] Laboratory investigation. J Neurosurg. 2010;112(4):868–879.
  • Yang D-Y, Sheu M-L, Su H-L, et al. Dual regeneration of muscle and nerve by intravenous administration of human amniotic fluid-derived mesenchymal stem cells regulated by stromal cell-derived factor-1α in a sciatic nerve injury model. J Neurosurg. 2012;116(6):1357–1367.
  • Su C-F, Chang L-H, Kao C-Y, et al. Application of amniotic fluid stem cells in repairing sciatic nerve injury in minipigs. Brain Res. 2018;1678:397–406.
  • Lemke A, Ferguson J, Gross K, et al. Transplantation of human amnion prevents recurring adhesions and ameliorates fibrosis in a rat model of sciatic nerve scarring. Acta Biomater. 2018;66:335–349.
  • Zarbakhsh S, Goudarzi N, Shirmohammadi M, et al. Histological study of bone marrow and umbilical cord stromal cell transplantation in regenerating rat peripheral nerve. Cell J (Yakhteh). 2016;17(4):668.
  • Li D, Wang C, Shan W, et al. Human amnion tissue injected with human umbilical cord mesenchymal stem cells repairs damaged sciatic nerves in rats. Neural Regen Res. 2012;7(23):1771.
  • Sung M-A, Jung HJ, Lee J-W, et al. Human umbilical cord blood-derived mesenchymal stem cells promote regeneration of crush-injured rat sciatic nerves. Neural Regen Res. 2012;7(26):2018.
  • Gärtner A, Pereira T, Armada-da-Silva P, et al. Use of poly (DL-lactide-ε-caprolactone) membranes and mesenchymal stem cells from the Wharton’s jelly of the umbilical cord for promoting nerve regeneration in axonotmesis: in vitro and in vivo analysis. Differentiation. 2012;84(5):355–365.
  • Gärtner A, Pereira T, Simões MJ, et al. Use of hybrid chitosan membranes and human mesenchymal stem cells from the Wharton jelly of umbilical cord for promoting nerve regeneration in an axonotmesis rat model. Neural Regen Res. 2012;7(29):2247.
  • Shalaby SM, Amal S, Ahmed FE, et al. Combined Wharton’s jelly-derived mesenchymal stem cells and nerve guidance conduit: A potential promising therapy for peripheral nerve injuries. Int J Biochem Cell Biol. 2017;86:67–76.
  • Snyder EY, Deitcher DL, Walsh C, et al. Multipotent neural cell lines can engraft and participate in development of mouse cerebellum. Cell. 1992;68(1):33–51.
  • Gonçalves JT, Schafer ST, Gage FH. Adult neurogenesis in the hippocampus: from stem cells to behavior. Cell. 2016;167(4):897–914.
  • Liard O, Segura S, Sagui E, et al. Adult-brain-derived neural stem cells grafting into a vein bridge increases postlesional recovery and regeneration in a peripheral nerve of adult pig. Stem Cells Int. 2012;2012:128732.
  • Ni H-C, Tseng T-C, Chen J-R, et al. Fabrication of bioactive conduits containing the fibroblast growth factor 1 and neural stem cells for peripheral nerve regeneration across a 15 mm critical gap. Biofabrication. 2013;5(3):035010.
  • Fu KY, Dai LG, Chiu IM, et al. Sciatic nerve regeneration by microporous nerve conduits seeded with glial cell line‐derived neurotrophic factor or brain‐derived neurotrophic factor gene transfected neural stem cells. Artif Organs. 2011;35(4):363–372.
  • Johnson TS, O’neill AC, Motarjem PM, et al. Tumor formation following murine neural precursor cell transplantation in a rat peripheral nerve injury model. J Reconstr Microsurg. 2008;24(8):545–550.
  • O’Rourke C, Day A, Murray-Dunning C, et al. An allogeneic ‘off the shelf’therapeutic strategy for peripheral nerve tissue engineering using clinical grade human neural stem cells. Sci Rep. 2018;8(1):2951.
  • Zhang Q, Nguyen P, Xu Q, et al. Neural progenitor‐like cells induced from human gingiva‐derived mesenchymal stem cells regulate myelination of Schwann cells in rat sciatic nerve regeneration. Stem Cells Transl Med. 2017;6(2):458–470.
  • Lee DC, Chen JH, Hsu TY, et al. Neural stem cells promote nerve regeneration through IL12-induced Schwann cell differentiation. Mol Cell Neurosci. 2017;79:1–11.
  • Strem BM, Hicok KC, Zhu M, et al. Multipotential differentiation of adipose tissue-derived stem cells. Keio J Med. 2005;54(3):132–141.
  • Sowa Y, Imura T, Numajiri T, et al. Adipose-derived stem cells produce factors enhancing peripheral nerve regeneration: influence of age and anatomic site of origin. Stem Cells Dev. 2011;21(11):1852–1862.
  • Nie C, Yang D, Xu J, et al. Locally administered adipose-derived stem cells accelerate wound healing through differentiation and vasculogenesis. Cell Transplant. 2011;20(2):205–216.
  • Faroni A, Smith RJ, Lu L, et al. Human Schwann‐like cells derived from adipose‐derived mesenchymal stem cells rapidly de‐differentiate in the absence of stimulating medium. Eur J Neurosci. 2016;43(3):417–430.
  • Tremp M, Sprenger L, Degrugillier L, et al. Regeneration of nerve crush injury using adipose-derived stem cells: A multimodal comparison. Muscle Nerve. 2018;58(4):566–572.
  • Bucan V, Vaslaitis D, Peck CT, et al. Effect of exosomes from rat adipose-derived mesenchymal stem cells on neurite outgrowth and sciatic nerve regeneration after crush injury. Mol Neurobiol. 2018. doi:10.1007/s12035-018-1172-z.
  • Sun X, Zhu Y, Yin HY, et al. Differentiation of adipose-derived stem cells into Schwann cell-like cells through intermittent induction: potential advantage of cellular transient memory function. Stem Cell Res Ther. 2018;9(1):133.
  • Fernandes M, Valente SG, Sabongi RG, et al. Bone marrow-derived mesenchymal stem cells versus adipose-derived mesenchymal stem cells for peripheral nerve regeneration. Neural Regen Res. 2018;13(1):100–104.
  • Allbright KO, Bliley JM, Havis E, et al. Delivery of adipose-derived stem cells in poloxamer hydrogel improves peripheral nerve regeneration. Muscle Nerve. 2018;58(2):251–260.
  • Masgutov R, Masgutova G, Mukhametova L, et al. Allogenic adipose-derived stem cells transplantation improved sciatic nerve regeneration in rats: autologous nerve graft model. Front Pharmacol. 2018;9:86.
  • Zhang M, Jiang MH, Kim DW, et al. Comparative analysis of the cell fates of induced Schwann cells from subcutaneous fat tissue and naive Schwann cells in the sciatic nerve injury model. Biomed Res Int. 2017;2017:1252851.
  • de Luca AC, Fonta CM, Raffoul W, et al. In vitro evaluation of gel-encapsulated adipose-derived stem cells: biochemical cues for in vivo peripheral nerve repair. J Tissue Eng Regen Med. 2018;12(3):676–686.
  • Hu F, Zhang X, Liu H, et al. Neuronally differentiated adipose-derived stem cells and aligned PHBV nanofiber nerve scaffolds promote sciatic nerve regeneration. Biochem Biophys Res Commun. 2017;489(2):171–178.
  • Haselbach D, Raffoul W, Larcher L, et al. di Summa PG. Regeneration patterns influence hindlimb automutilation after sciatic nerve repair using stem cells in rats. Neurosci Lett. 2016;634:153–159.
  • Cherubino M, Pellegatta I, Crosio A, et al. Use of human fat grafting in the prevention of perineural adherence: experimental study in athymic mouse. PLoS One. 2017;12(4):e0176393.
  • Hernandez-Cortes P, Toledo-Romero MA, Delgado M, et al. Ghrelin and adipose-derived mesenchymal stromal cells improve nerve regeneration in a rat model of epsilon-caprolactone conduit reconstruction. Histol Histopathol. 2017;32(6):627–637.
  • Hu Y, Wu Y, Gou Z, et al. 3D-engineering of cellularized conduits for peripheral nerve regeneration. Sci Rep. 2016;6:32184.
  • He X, Ao Q, Wei Y, et al. Transplantation of miRNA-34a overexpressing adipose-derived stem cell enhances rat nerve regeneration. Wound Repair Regen. 2016;24(3):542–550.
  • Sowa Y, Kishida T, Imura T, et al. Adipose-derived stem cells promote peripheral nerve regeneration in vivo without differentiation into Schwann-like lineage. Plast Reconstr Surg. 2016;137(2):318e–30e.
  • Cuevas P, Carceller F, Dujovny M, et al. Peripheral nerve regeneration by bone marrow stromal cells. Neurol Res. 2002;24(7):634–638.
  • Lu D, Mahmood A, Wang L, et al. Adult bone marrow stromal cells administered intravenously to rats after traumatic brain injury migrate into brain and improve neurological outcome. Neuroreport. 2001;12(3):559–563.
  • Nakano‐Doi A, Nakagomi T, Fujikawa M, et al. Bone marrow mononuclear cells promote proliferation of endogenous neural stem cells through vascular niches after cerebral infarction. Stem Cells. 2010;28(7):1292–1302.
  • Kaka G, Arum J, Sadraie SH, et al. Bone marrow stromal cells associated with Poly L-Lactic-Co-Glycolic Acid (PLGA) nanofiber scaffold improve transected sciatic nerve regeneration. Iran J Biotechnol. 2017;15(3):149–156.
  • Zheng Y, Huang C, Liu F, et al. Reactivation of denervated Schwann cells by neurons induced from bone marrow-derived mesenchymal stem cells. Brain Res Bull. 2018;139:211–223.
  • Ozer H, Bozkurt H, Bozkurt G, et al. Regenerative potential of chitosan-coated poly-3-hydroxybutyrate conduits seeded with mesenchymal stem cells in a rat sciatic nerve injury model. Int J Neurosci. 2018;128(9):828–834.
  • Kizilay Z, Aktas S, Kahraman Cetin N, et al. Effect of systemic application of bone marrow-derived mesenchymal stem cells on healing of peripheral nerve injury in an experimental sciatic nerve injury model. Turk Neurosurg. 2017;28(4):654–662.
  • Cai S, Tsui YP, Tam KW, et al. Directed differentiation of human bone marrow stromal cells to fate-committed Schwann cells. Stem Cell Reports. 2017;9(4):1097–1108.
  • Zheng M, Duan J, He Z, et al. Transplantation of bone marrow stromal stem cells overexpressing tropomyosin receptor kinase A for peripheral nerve repair. Cytotherapy. 2017;19(8):916–926.
  • Garcia-Perez MM, Martinez-Rodriguez HG, Lopez-Guerra GG, et al. A modified chemical protocol of decellularization of rat sciatic nerve and its recellularization with mesenchymal differentiated Schwann-like cells: morphological and functional assessments. Histol Histopathol. 2017;32(8):779–792.
  • Wang Y, Jia H, Li WY, et al. Molecular examination of bone marrow stromal cells and chondroitinase ABC-assisted acellular nerve allograft for peripheral nerve regeneration. Exp Ther Med. 2016;12(4):1980–1992.
  • Kaizawa Y, Kakinoki R, Ikeguchi R, et al. A nerve conduit containing a vascular bundle and implanted with bone marrow stromal cells and decellularized allogenic nerve matrix. Cell Transplant. 2017;26(2):215–228.
  • Cooney DS, Wimmers EG, Ibrahim Z, et al. Mesenchymal stem cells enhance nerve regeneration in a rat sciatic nerve repair and hindlimb transplant model. Sci Rep. 2016;6:31306.
  • Gu Y, Li Z, Huang J, et al. Application of marrow mesenchymal stem cell-derived extracellular matrix in peripheral nerve tissue engineering. J Tissue Eng Regen Med. 2017;11(8):2250–2260.
  • Zhang YR, Ka K, Zhang GC, et al. Repair of peripheral nerve defects with chemically extracted acellular nerve allografts loaded with neurotrophic factors-transfected bone marrow mesenchymal stem cells. Neural Regen Res. 2015;10(9):1498–1506.
  • Ke X, Li Q, Xu L, et al. Netrin-1 overexpression in bone marrow mesenchymal stem cells promotes functional recovery in a rat model of peripheral nerve injury. J Biomed Res. 2015;29(5):380–389.
  • Urish KL, Vella JB, Okada M, et al. Antioxidant levels represent a major determinant in the regenerative capacity of muscle stem cells. Mol Biol Cell. 2009;20(1):509–520.
  • Tamaki T, Hirata M, Soeda S, et al. Preferential and comprehensive reconstitution of severely damaged sciatic nerve using murine skeletal muscle-derived multipotent stem cells. PLoS One. 2014;9(3):e91257.
  • Tamaki T, Hirata M, Nakajima N, et al. A long-gap peripheral nerve injury therapy using human skeletal muscle-derived stem cells (Sk-SCs): an achievement of significant morphological, numerical and functional recovery. PLoS One. 2016;11(11):e0166639.
  • Lavasani M, Thompson SD, Pollett JB, et al. Human muscle-derived stem/progenitor cells promote functional murine peripheral nerve regeneration. J Clin Invest. 2014;124(4):1745–1756.
  • Seta H, Maki D, Kazuno A, et al. Voluntary exercise positively affects the recovery of long-nerve gap injury following tube-bridging with human skeletal muscle-derived stem cell transplantation. J Clin Med. 2018;7(4):67.
  • Amoh Y, Li L, Campillo R, et al. Implanted hair follicle stem cells form Schwann cells that support repair of severed peripheral nerves. Proc Nat Acad Sci. 2005;102(49):17734–17738.
  • Lin H, Liu F, Zhang C, et al. Characterization of nerve conduits seeded with neurons and Schwann cells derived from hair follicle neural crest stem cells. Tissue Eng Part A. 2011;17(13–14):1691–1698.
  • Yamazaki A, Obara K, Tohgi N, et al. Implanted hair-follicle-associated pluripotent (HAP) stem cells encapsulated in polyvinylidene fluoride membrane cylinders promote effective recovery of peripheral nerve injury. Cell Cycle (Georgetown, Tex). 2017;16(20):1927–1932.
  • Martens W, Sanen K, Georgiou M, et al. Human dental pulp stem cells can differentiate into Schwann cells and promote and guide neurite outgrowth in an aligned tissue-engineered collagen construct in vitro. FASEB J. 2014;28(4):1634–1643.
  • Kolar MK, Itte VN, Kingham PJ, et al. The neurotrophic effects of different human dental mesenchymal stem cells. Sci Rep. 2017;7(1):12605.
  • Ullah I, Park J-M, Kang Y-H, et al. Transplantation of human dental pulp-derived stem cells or differentiated neuronal cells from human dental pulp-derived stem cells identically enhances regeneration of the injured peripheral nerve. Stem Cells Dev. 2017;26(17):1247–1257.
  • Sanen K, Martens W, Georgiou M, et al. Engineered neural tissue with Schwann cell differentiated human dental pulp stem cells: potential for peripheral nerve repair? J Tissue Eng Regen Med. 2017;11(12):3362–3372.
  • Carnevale G, Pisciotta A, Riccio M, et al. Human dental pulp stem cells expressing STRO‐1, c‐kit and CD34 markers in peripheral nerve regeneration. J Tissue Eng Regen Med. 2018;12(2):e774–e85.
  • Askari N, Yaghoobi M, Shamsara M, et al. Tetracycline-regulated expression of OLIG2 gene in human dental pulp stem cells lead to mouse sciatic nerve regeneration upon transplantation. Neuroscience. 2015;305:197–208.
  • Yamamoto T, Osako Y, Ito M, et al. Trophic effects of dental pulp stem cells on Schwann cells in peripheral nerve regeneration. Cell Transplant. 2016;25(1):183–193.
  • McKenzie IA, Biernaskie J, Toma JG, et al. Skin-derived precursors generate myelinating Schwann cells for the injured and dysmyelinated nervous system. J Neurosci. 2006;26(24):6651–6660.
  • Shakhbazau A, Mohanty C, Kumar R, et al. Sensory recovery after cell therapy in peripheral nerve repair: effects of naive and skin precursor-derived Schwann cells. J Neurosurg. 2014;121(2):423–431.
  • Stratton JA, Shah PT, Kumar R, et al. The immunomodulatory properties of adult skin‐derived precursor Schwann cells: implications for peripheral nerve injury therapy. Eur J Neurosci. 2016;43(3):365–375.
  • Khuong HT, Kumar R, Senjaya F, et al. Skin-derived precursor Schwann cells improve behavioral recovery for acute and delayed nerve repair. Exp Neurol. 2014;254:168–179.
  • Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126(4):663–676.
  • Wang A, Tang Z, Park I-H, et al. Induced pluripotent stem cells for neural tissue engineering. Biomaterials. 2011;32(22):5023–5032.
  • Uemura T, Ikeda M, Takamatsu K, et al. Long-term efficacy and safety outcomes of transplantation of induced pluripotent stem cell-derived neurospheres with bioabsorbable nerve conduits for peripheral nerve regeneration in mice. Cells Tissues Organs. 2014;200(1):78–91.
  • Ikeda M, Uemura T, Takamatsu K, et al. Acceleration of peripheral nerve regeneration using nerve conduits in combination with induced pluripotent stem cell technology and a basic fibroblast growth factor drug delivery system. J Biomed Mater Res A. 2014;102(5):1370–1378.
  • Huang C-W, Huang W-C, Qiu X, et al. The differentiation stage of transplanted stem cells modulates nerve regeneration. Sci Rep. 2017;7(1):17401.
  • Kimura H, Ouchi T, Shibata S, et al. Stem cells purified from human induced pluripotent stem cell-derived neural crest-like cells promote peripheral nerve regeneration. Sci Rep. 2018;8(1):10071.
  • Muhammad A, Kim K, Epifantseva I, et al. Cell transplantation strategies for acquired and inherited disorders of peripheral myelin. Ann Clin Transl Neurol. 2018;5(2):186–200.
  • Pepper J-P, Wang TV, Hennes V, et al. Human induced pluripotent stem cell-derived motor neuron transplant for neuromuscular atrophy in a mouse model of sciatic nerve injury. JAMA Facial Plast Surg. 2017;19(3):197–205.
  • Lv Y, Nan P, Chen G, et al. In vivo repair of rat transected sciatic nerve by low-intensity pulsed ultrasound and induced pluripotent stem cells-derived neural crest stem cells. Biotechnol Lett. 2015;37(12):2497–2506.
  • Yokoi T, Uemura T, Takamatsu K, et al. Bioabsorbable nerve conduits coated with induced pluripotent stem cell‐derived neurospheres enhance axonal regeneration in sciatic nerve defects in aged mice. J Biomed Mater Res Part B. 2018;106(5):1752–1758.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.