Journal Home > Volume 5 , Issue 1
Background:

To investigate the biocompatibility of electrospinning polycaprolactone (PCL) fiber scaffolds and coculture system, which consisted of neural stem cells (NSCs) and activated Schwann cells (ASCs).

Materials and methods:

ASCs were isolated from sciatic nerves, ligated for 7 days, in 4-week-old Wistar rats, and the NSCs were isolated from the hippocampus of E14.5 Wistar rat embryos. ASCs, NSCs and ASCs combined with NSCs were 3D cultured on the electrospinning PCL fiber scaffolds. Crystal violet staining was used to find the suitable density of ASCs for growth, and the proliferation of NSCs and ASCs were tested by Cell Counting Kit (CCK)-8 assay, and cell adhesion, differentiation of NSCs and myelin basic protein (MBP) expression of ASCs were observed by laser confocal microscopy. Distribution and morphology were assessed by scanning electron microscopy.

Results:

The average diameter of fibers in electrospinning PCL scaffolds was approximately 7.93±1.41 μm. ASCs could grow well at the density of 2×104/cm2, and a certain number of cells extended along the longitudinal axis of fibers, and the shape of the cells was spindle, which was consistent with crystal violet staining results. The CCK-8 experiment showed ASCs could proliferate gradually on the PCL scaffold within 7 days, as well as NSCs, and NSCs differentiated into astrocytes, neurons and oligodendrocytes on the PCL scaffold; PCL scaffolds could improve the differentiation rate of neurons. After NSCs and ASCs were cocultured on electrospinning PCL scaffolds, ASCs could express MBP and NSCs could differentiate into neurons, which distributed around those ASCs expressing MBP.

Conclusion:

Electrospinning PCL fibrous scaffolds showed good biocompatibility, and the fibers had an inducing effect on the distribution of ASCs. NSCs and ASCs cultured on electrospinning PCL scaffolds could form 3D culture system, and NSCs could differentiate into neurons which distributed around the ASCs expressing MBP.


menu
Abstract
Full text
Outline
About this article

In vitro study of neural stem cells and activated Schwann cells cocultured on electrospinning polycaprolactone scaffolds

Show Author's information Baoyou Fan1,2,*Xianhu Zhou1,2,*Lina Wang3,*Zhijian Wei1,2Wei Lin1,2Yiming Ren1,2Guidong Shi1,2Xin Cheng1,2Lianyong Wang3( )Shiqing Feng1,2( )
International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedic Surgery, Tianjin Medical University General Hospital,
Tianjin Neurological Institute, Key Laboratory of Post-neuroinjury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City,
Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, People’s Republic of China

*These authors contributed equally to this work

Abstract

Background:

To investigate the biocompatibility of electrospinning polycaprolactone (PCL) fiber scaffolds and coculture system, which consisted of neural stem cells (NSCs) and activated Schwann cells (ASCs).

Materials and methods:

ASCs were isolated from sciatic nerves, ligated for 7 days, in 4-week-old Wistar rats, and the NSCs were isolated from the hippocampus of E14.5 Wistar rat embryos. ASCs, NSCs and ASCs combined with NSCs were 3D cultured on the electrospinning PCL fiber scaffolds. Crystal violet staining was used to find the suitable density of ASCs for growth, and the proliferation of NSCs and ASCs were tested by Cell Counting Kit (CCK)-8 assay, and cell adhesion, differentiation of NSCs and myelin basic protein (MBP) expression of ASCs were observed by laser confocal microscopy. Distribution and morphology were assessed by scanning electron microscopy.

Results:

The average diameter of fibers in electrospinning PCL scaffolds was approximately 7.93±1.41 μm. ASCs could grow well at the density of 2×104/cm2, and a certain number of cells extended along the longitudinal axis of fibers, and the shape of the cells was spindle, which was consistent with crystal violet staining results. The CCK-8 experiment showed ASCs could proliferate gradually on the PCL scaffold within 7 days, as well as NSCs, and NSCs differentiated into astrocytes, neurons and oligodendrocytes on the PCL scaffold; PCL scaffolds could improve the differentiation rate of neurons. After NSCs and ASCs were cocultured on electrospinning PCL scaffolds, ASCs could express MBP and NSCs could differentiate into neurons, which distributed around those ASCs expressing MBP.

Conclusion:

Electrospinning PCL fibrous scaffolds showed good biocompatibility, and the fibers had an inducing effect on the distribution of ASCs. NSCs and ASCs cultured on electrospinning PCL scaffolds could form 3D culture system, and NSCs could differentiate into neurons which distributed around the ASCs expressing MBP.

Keywords: electrospinning, Schwann cells, neural stem cells, scaffold, PCL

References(46)

1.
Mehrabi S, Eftekhari S, Moradi F, et al. Cell therapy in spinal cord injury: a mini-review. Basic Clin Neurosci. 2013;4(2):172–176.
2.
Barnabé-Heider F, Frisén J. Stem cells for spinal cord repair. Cell Stem Cell. 2008;3(1):16–24.
3.
Tremp M, Meyer Zu Schwabedissen M, Kappos EA, et al. The regeneration potential after human and autologous stem cell transplantation in a rat sciatic nerve injury model can be monitored by MRI. Cell Transplant. 2015;24(2):203–211.
4.
Wei L, Wei ZZ, Jiang MQ, Mohamad O, Yu SP. Stem cell transplantation therapy for multifaceted therapeutic benefits after stroke. Prog Neurobiol. Epub 2017 Mar 18.
5.
Ronaghi M, Erceg S, Moreno-Manzano V, Stojkovic M. Challenges of stem cell therapy for spinal cord injury: human embryonic stem cells, endogenous neural stem cells, or induced pluripotent stem cells? Stem Cells. 2010;28(1):93–99.
6.
Wiliams RR, Bunge MB. Schwann cell transplantation: a repair strategy for spinal cord injury? Prog Brain Res. 2012;201:295–312.
7.
Silva NA, Sousa N, Reis RL, Salgado AJ. From basics to clinical: a comprehensive review on spinal cord injury. Prog Neurobiol. 2014;114:25–57.
8.
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.
9.
Barnett SC, Riddell JS. Olfactory ensheathing cells (OECs) and the treatment of CNS injury: advantages and possible caveats. J Anat. 2004;204(1):57–67.
10.
Liang P, Liu J, Xiong J, et al. Neural stem cell-conditioned medium protects neurons and promotes propriospinal neurons relay neural circuit reconnection after spinal cord injury. Cell Transplant. 2014;23 (Suppl 1):S45–S56.
11.
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(6):1264–1273.
12.
Sabelström H, Stenudd M, Réu P, et al. Resident neural stem cells restrict tissue damage and neuronal loss after spinal cord injury in mice. Science. 2013;342(6158):637–640.
13.
Tao Li J, Somasundaram C, Bian K, et al. Nitric oxide signaling and neural stem cell differentiation in peripheral nerve regeneration. Eplasty. 2010;10:e42.
14.
Dadon-Nachum M, Melamed E, Offen D. Stem cells treatment for sciatic nerve injury. Expert Opin Biol Ther. 2011;11(12):1591–1597.
15.
Piltti KM, Avakian SN, Funes GM, et al. Transplantation dose alters the dynamics of human neural stem cell engraftment, proliferation and migration after spinal cord injury. Stem Cell Res. 2015;15(2):341–353.
16.
Song S, Kamath S, Mosquera D, et al. Expression of brain natriuretic peptide by human bone marrow stromal cells. Exp Neurol. 2004;185(1):191–197.
17.
Wu S, Suzuki Y, Kitada M, et al. Migration, integration, and differentiation of hippocampus-derived neurosphere cells after transplantation into injured rat spinal cord. Neurosci Lett. 2001;312(3):173–176.
18.
Houweling DA, Lankhorst AJ, Gispen WH, Bär PR, Joosten EA. Collagen containing neurotrophin-3 (NT-3) attracts regrowing injured corticospinal axons in the adult rat spinal cord and promotes partial functional recovery. Exp Neurol. 1998;153(1):49–59.
19.
Enomoto M. Therapeutic effects of neurotrophic factors in experimental spinal cord injury models. J Neurorestoratol. 2016;4(1):15–22.
20.
Fon D, Zhou K, Ercole F, et al. Nanofibrous scaffolds releasing a small molecule BDNF-mimetic for the re-direction of endogenous neuroblast migration in the brain. Biomaterials. 2014;35(9):2692–2712.
21.
Akassoglou K, Yu WM, Akpinar P, Strickland S. Fibrin inhibits peripheral nerve remyelination by regulating Schwann cell differentiation. Neuron. 2002;33(6):861–875.
22.
Keilhoff G, Fansa H, Schneider W, Wolf G. In vivo predegeneration of peripheral nerves: an effective technique to obtain activated Schwann cells for nerve conduits. J Neurosci Methods. 1999;89(1):17–24.
23.
Pearse DD, Barakat DJ. Cellular repair strategies for spinal cord injury. Expert Opin Biol Ther. 2006;6(7):639–652.
24.
Wang JM, Zeng YS, Wu JL, Li Y, Teng YD. Cograft of neural stem cells and schwann cells overexpressing TrkC and neurotrophin-3 respectively after rat spinal cord transection. Biomaterials. 2011;32(30):7454–7468.
25.
Yang Y, Kamudzandu M, Roach P, Fricker R. Nanofibrous scaffolds supporting optimal central nervous system regeneration: an evidence-based review. J Neurorestoratol. 2015;3:123–131.
26.
Middleton JC, Tipton AJ. Synthetic biodegradable polymers as orthopedic devices. Biomaterials. 2000;21(23):2335–2346.
27.
Ng KW, Achuth HN, Moochhala S, Lim TC, Hutmacher DW. In vivo evaluation of an ultra-thin polycaprolactone film as a wound dressing. J Biomater Sci Polym Ed. 2007;18(7):925–938.
28.
Woodruff MA, Hutmacher DW. The return of a forgotten polymer—polycaprolactone in the 21st century. Prog Polym Sci. 2010;35(10):1217–1256.
29.
Kim DY, Choi YS, Kim SE, et al. In vivo effects of adipose-derived stem cells in inducing neuronal regeneration in Sprague-Dawley rats undergoing nerve defect bridged with polycaprolactone nanotubes. J Korean Med Sci. 2014;29 (Suppl 3):S183–S192.
30.
Donoghue PS, Lamond R, Boomkamp SD, et al. The development of a ε-polycaprolactone scaffold for central nervous system repair. Tissue Eng Part A. 2013;19(3–4):497–507.
31.
Beigi MH, Ghasemi-Mobarakeh L, Prabhakaran MP, et al. In vivo integration of poly(ε-caprolactone)/gelatin nanofibrous nerve guide seeded with teeth derived stem cells for peripheral nerve regeneration. J Biomed Mater Res A. 2014;102(12):4554–4567.
32.
Azari H, Sharififar S, Rahman M, Ansari S, Reynolds BA. Establishing embryonic mouse neural stem cell culture using the neurosphere assay. J Vis Exp. 2011;(47):pii 2457.
33.
Cho K-S, Pearse DD, Deitrich DW, et al. Schwann cell transplantation improves reticulo-spinal fiber growth and forelimb strength after severe cervical spinal cord contusion. Exp Neurol. 2007;193:239.
34.
Hill CE, Hurtado A, Blits B, et al. Early necrosis and apoptosis of Schwann cells transplanted into the injured rat spinal cord. Eur J Neurosci. 2007;26(6):1433–1445.
35.
Barakat DJ, Gaglani SM, Neravetla SR, et al. Survival, integration, and axon growth support of glia transplanted into the chronically contused spinal cord. Cell Transplant. 2005;14(4):225–240.
36.
Pearse DD, Sanchez AR, Pereira FC, et al. Transplantation of Schwann cells and/or olfactory ensheathing glia into the contused spinal cord: survival, migration, axon association, and functional recovery. Glia. 2007;55(9):976–1000.
37.
Niapour A, Karamali F, Nemati S, et al. Cotransplantation of human embryonic stem cell-derived neural progenitors and schwann cells in a rat spinal cord contusion injury model elicits a distinct neurogenesis and functional recovery. Cell Transplant. 2012;21(5):827–843.
38.
Wang X, Xu XM. Long-term survival, axonal growth-promotion, and myelination of Schwann cells grafted into contused spinal cord in adult rats. Exp Neurol. 2014;261:308–319.
39.
Bunge MB. Efficacy of Schwann cell transplantation for spinal cord repair is improved with combinatorial strategies. J Physiol. 2016;594(13):3533–3538.
40.
Thompson DM, Buettner HM. Neurite outgrowth is directed by schwann cell alignment in the absence of other guidance cues. Ann Biomed Eng. 2006;34(1):161–168.
41.
Xiang L, Chen Y. Stem cell transplantation for treating spinal cord injury: a literature comparison between studies of stem cells obtained from various sources. Neural Regen Res. 2012;7(16):1256–1263.
42.
Sørensen A, Alekseeva T, Katechia K, Robertson M, Riehle MO, Barnett SC. Long-term neurite orientation on astrocyte monolayers aligned by microtopography. Biomaterials. 2007;28(36):5498–5508.
43.
Ebrahimi-Barough S, Hoveizi E, Yazdankhah M, et al. Inhibitor of PI3K/Akt signaling pathway small molecule promotes motor neuron differentiation of human endometrial stem cells cultured on electrospun biocomposite polycaprolactone/collagen scaffolds. Mol Neurobiol. 2016;54(4):2547–2554.
44.
Raspa A, Marchini A, Pugliese R, et al. A biocompatibility study of new nanofibrous scaffolds for nervous system regeneration. Nanoscale. 2016;8(1):253–265.
45.
Wang J, Ye R, Wei Y, et al. The effects of electrospun TSF nanofiber diameter and alignment on neuronal differentiation of human embryonic stem cells. J Biomed Mater Res A. 2012;100(3):632–645.
46.
Kamei KI, Koyama Y, Tokunaga Y, et al. Characterization of phenotypic and transcriptional differences in human pluripotent stem cells under 2D and 3D culture conditions. Adv Healthc Mater. 2016;5(22):2951–2958.
Publication history
Copyright
Acknowledgements
Rights and permissions

Publication history

Published: 06 September 2017
Issue date: December 2017

Copyright

© 2017 The Author(s).

Acknowledgements

This study was supported by the State Key Program of National Natural Science Foundation of China (81330042), Special Program for Sino-Russian Joint Research Sponsored by the Ministry of Science and Technology, China (2014DFR31210), International Cooperation Program of National Natural Science Foundation of China (81620108018), and Key Program Sponsored by the Tianjin Science and Technology Committee, China (14ZCZDSY00044, 13RCGFSY19000). The authors also thank Yan Hao and Baowen Liu for their help with the experiment.

Rights and permissions

© 2017 Fan et al. This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License (http://creativecommons.org/licenses/by-nc/3.0/). By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms (https://www.dovepress.com/terms.php).

Return