リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

大学・研究所にある論文を検索できる 「Transplantation of rat cranial bone-derived mesenchymal stem cells promotes functional recovery in rats with spinal cord injury」の論文概要。リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

コピーが完了しました

URLをコピーしました

論文の公開元へ論文の公開元へ
書き出し

Transplantation of rat cranial bone-derived mesenchymal stem cells promotes functional recovery in rats with spinal cord injury

前田 雄洋 広島大学

2022.03.23

概要

www.nature.com/scientificreports

OPEN

Transplantation of rat cranial
bone‑derived mesenchymal stem
cells promotes functional recovery
in rats with spinal cord injury
Yuyo Maeda1*, Takashi Otsuka2, Masaaki Takeda1, Takahito Okazaki1, Kiyoharu Shimizu1,
Masashi Kuwabara1, Masahiro Hosogai1, Louis Yuge2 & Takafumi Mitsuhara1
Cell-based therapy using mesenchymal stem cells (MSCs) is a novel treatment strategy for spinal
cord injury (SCI). MSCs can be isolated from various tissues, and their characteristics vary based
on the source. However, reports demonstrating the effect of transplanted rat cranial bone-derived
MSCs (rcMSCs) on rat SCI models are lacking. In this study, we determined the effect of transplanting
rcMSCs in rat SCI models. MSCs were established from collected bone marrow and cranial bones. SCI
rats were established using the weight-drop method and transplanted intravenously with MSCs at
24 h post SCI. The recovery of motor function and hindlimb electrophysiology was evaluated 4 weeks
post transplantation. Electrophysiological recovery was evaluated by recording the transcranial
electrical stimulation motor-evoked potentials. Tissue repair after SCI was assessed by calculating
the cavity ratio. The expression of genes involved in the inflammatory response and cell death in the
spinal cord tissue was assessed by real-time polymerase chain reaction. The transplantation of rcMSCs
improved motor function and electrophysiology recovery, and reduced cavity ratio. The expression of
proinflammatory cytokines was suppressed in the spinal cord tissues of the rats that received rcMSCs.
These results demonstrate the efficacy of rcMSCs as cell-based therapy for SCI.
Spinal cord injury (SCI) can cause severe damage, leading to permanent loss of mobility, incontinence, and other
functional losses. In the absence of effective treatments for SCI, surgical restabilization of the vertebral column
and rehabilitation are currently the primary therapeutic o
­ ptions1. Cell-based therapy using mesenchymal stem
cells (MSCs) has garnered attention as a novel approach for treating the damage caused by SCI. MSCs can be
isolated from various tissues, such as bone m
­ arrow2 or adipose t­ issue3, and possess self-renewal and multilineage differentiation potentials. Some studies have revealed that the characteristics of MSCs may vary in these
­tissues4–6. In a previous study, researchers successfully established MSCs from rat and human cranial bones,
and demonstrated that they secrete abundant neurotrophic factors when transplanted in a rat model of cerebral
­infarction7,8. However, reports demonstrating the effect of transplanted cranial bone-derived MSCs on rat SCI
models are lacking. In addition, the mechanism of action of cranial bone-derived MSCs in central nervous
system (CNS) disorders has not yet been i­nvestigated7–9. Motor function recovery has been evaluated only via
behavioral assessments, as the electrophysiological recovery has not been determined. Furthermore, no studies
have investigated the effect of transplanting MSCs on neurophysiological recovery over time using extended
neurophysiological evaluation. We have previously established techniques for recording extended transcranial
electrical stimulation motor-evoked potentials (tcMEPs) in r­ ats10.
Therefore, in the present study, we investigated the effects of transplanting rat cranial bone-derived MSCs
(rcMSCs) on the neurophysiology of an SCI rat model. Moreover, we determined the neurophysiological recovery
using extended tcMEP recordings and the mechanism underlying rcMSCs function in an SCI model. We propose
rcMSCs as a potential new source of cell therapy against SCIs.

1

Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Kasumi
1‑2‑3, Minami‑ku, Hiroshima, Japan. 2Division of Bio‑Environmental Adaptation Sciences, Graduate School of
Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan. *email: d182072@hiroshima-u.ac.jp

Scientific Reports |

(2021) 11:21907

| https://doi.org/10.1038/s41598-021-01490-1

1
Vol.:(0123456789)

www.nature.com/scientificreports/

Results

Flow cytometry analysis for MSC‑specific markers.  MSC-specific markers in rat bone marrow mesenchymal stem cells (rbMSCs) and rcMSCs were analyzed using flow cytometry. The results were positive for
cell surface markers associated with MSCs, such as CD29, CD90, and CD44 (Supplementary Table S4 online);
in contrast, they were negative for cell surface markers associated with hematopoietic cells, such as CD34 and
CD45 (Supplementary Table S4 online). rbMSCs and rcMSCs exhibited similar characteristics, with their cell
surface markers resembling those described in previous ­reports7–9.
Multilineage cell differentiation.  The differentiation potentials of the isolated rbMSCs and rcMSCs into

osteogenic and adipocytic cells were investigated. Neither type of MSCs exhibited positive staining with Alizarin
red S before osteogenic differentiation; however, positive cells were observed after differentiation (Supplementary Fig. S1A online), whereas staining with oil red O revealed that both types of MSCs tended to differentiate
into adipocytic cells (Supplementary Fig. S1 online).

Analysis of the genetic landscape of MSCs.  The expression of genes encoding neurotrophic factors

and anti-inflammatory factors in rbMSCs and rcMSCs was analyzed using real-time polymerase chain reaction
(PCR). The expression of Bdnf, Gdnf, Ngf, and Vegf, which encode neurotrophic factors, and that of Tgfb and
Tsg6, which encode anti-inflammatory factors, was evaluated. The expression of Bdnf, Gdnf, and Vegf was significantly higher in rcMSCs than in rbMSCs (P < 0.05, P < 0.05, P < 0.05, respectively; Fig. 1a,b,d). The expression of
Ngf and of genes encoding anti-inflammatory factors did not differ significantly between rcMSCs and rbMSCs
at passage 3 (Fig. 1c,e,f).

Effects of cell transplantation in an SCI rat model: recovery of motor function.  We assessed the
neurological function using the Basso–Beattie–Bresnahan (BBB) scale, and the inclined plane task score was
used to assess the functional benefits after the transplantation of rbMSCs and rcMSCs post-SCI. All rats showed
recovery of motor function in these tests after SCI. Improvements in the BBB scale score were significantly
greater in the rcMSC group than those in the phosphate-buffered saline (PBS) group on day 1 (P < 0.05; Fig. 2a)
and on days 3, 5, 7, 10, 14, 21, and 28 after injury (P < 0.01; Fig. 2a). In addition, improvements in the rcMSC
group were also higher than those in the rbMSC groups on day 3 (P < 0.05; Fig. 2a) and on days 5, 7, 10, 14, 21,
and 28 after injury (P < 0.01; Fig. 2a). Improvements in inclined plane task scores were significantly higher in the
rcMSC group than in the PBS group at 2, 3, 5, 7, 10, 14, 21, and 28 days after injury (P < 0.01; Fig. 2b). In addition,
improvements in the rcMSC group were also higher than those in the rbMSC group on day 2 (P < 0.05; Fig. 2b)
and on days 3, 5, 7, 10, 14, 21, and 28 after injury (P < 0.01; Fig. 2b). Rats in the rcMSC group exhibited more
motor function recovery than those in the other groups.
Effects of cell transplantation in an SCI rat model: electrophysiological recovery.  Representa-

tive waveforms of tcMEPs pre-operation and at 1, 7, 21, and 28 days after SCI are shown in Fig. 3a–c. The waveform of tcMEPs disappeared and gradually recovered after SCI. The recovery rate of the amplitude improved
at 14, 21, and 28 days after SCI compared with that on the day after injury in the rbMSC and rcMSC groups
(Fig. 3d). In addition, the recovery rate of the amplitudes in the rcMSC group was higher than that in other
groups on days 14 and 21 (P < 0.05; Fig. 3d) and at 28 days after SCI (P < 0.01; Fig. 3d).

Effects of cell transplantation in an SCI rat model: cavity repair.  We assessed cavity repair using
the cavity ratio and compared the histological recovery after the transplantation of rbMSCs and rcMSCs following SCI. Only small cavities were identified within the SCI lesions in the rcMSC group (Fig. 4a–d). The cavity
ratios of the rbMSC and rcMSC groups were significantly lower than those of the PBS group (Fig. 4e). The cavity
ratio of the rcMSC group was also significantly lower than that of the rbMSC group (P < 0.05; Fig. 4e).
Spinal cord tissue sampling and mRNA expression analysis of the spinal cord lesion site.  The
mRNA expression of Bax/Bcl2 and Casp3/Gapdh did not differ significantly between the three groups (Fig. 5a,b).
Moreover, the levels of Il1b were significantly lower in the rbMSC and rcMSC groups than in the PBS group
(P < 0.05; Fig. 5c), and those of Tnfa were significantly lower in the rcMSC group than in the PBS group (P < 0.05;
Fig. 5d).
NG108‑15 cell death assay after exposure to oxidative and inflammatory stress.  The survival
rate of NG108-15 cells exposed to inflammatory stress and cultured in rcMSC-conditioned medium (CM) was
significantly higher than that in the other groups (P < 0.01; Fig. 6a). Moreover, the survival rate of NG108-15
cells exposed to oxidative stress and cultured in rcMSC-CM or rbMSC-CM was significantly higher than that in
control cells (P < 0.01 and P < 0.05, respectively; Fig. 6b).
Analysis of mRNA expression in stress‑exposed NG108‑15 cells.  The Bax/Bcl2 ratio and Casp3
expression in inflammatory stress-exposed NG108-15 cells did not differ significantly between the three groups
(Fig. 7a,b), whereas they were significantly lower in oxidative stress-exposed NG108-15 cells cultured in rcMSCCM than in control cells (P < 0.05 and P < 0.01, respectively; Fig. 7c,d). The Bax/Bcl2 ratio in cells cultured in
rbMSC-CM was also significantly lower than that in control cells (P < 0.05; Fig. 7c). ...

この論文で使われている画像

参考文献

1. Muniswami, D. M., Kanthakumar, P., Kanakasabapathy, I. & Tharion, G. Motor recovery after transplantation of bone marrow

mesenchymal stem cells in rat models of spinal cord injury. Ann. Neurosci. 25, 126–140 (2019).

2. Pittenger, M. F. et al. Multilineage potential of adult human mesenchymal stem cells. Science 284, 143–147 (1999).

3. Zuk, P. A. et al. Human adipose tissue is a source of multipotent stem cells. Mol. Biol. Cell. 13, 4279–4295 (2002).

4. Kisiel, A. H. et al. Isolation, characterization, and in vitro proliferation of canine mesenchymal stem cells derived from bone marrow, adipose tissue, muscle, and periosteum. Am. J. Vet. Res. 73, 1305–1317 (2012).

5. Reich, C. M. et al. Isolation, culture and chondrogenic differentiation of canine adipose tissue- and bone marrow-derived mesenchymal stem cells: A comparative study. Vet. Res. Commun. 36, 139–148 (2012).

6. Melief, S. M., Jan Zwaginga, J., Fibbe, W. E. & Roelofs, H. Adipose tissue-derived multipotent stromal cells have a higher immunomodulatory capacity than their bone marrow-derived counterparts. Stem Cells Transl. Med. 2, 455–463 (2013).

7. Abiko, M. et al. Rat cranial bone-derived mesenchymal stem cell transplantation promotes functional recovery in ischemic stroke

model rats. Stem Cells Dev. 27, 1053–1061 (2018).

8. Oshita, J. et al. Early transplantation of human cranial bone-derived mesenchymal stem cells enhances functional recovery in

ischemic stroke model rats. Neurol. Med. Chir. 60, 83–93 (2020).

9. Shinagawa, K. et al. The characteristics of human cranial bone marrow mesenchymal stem cells. Neurosci. Lett. 606, 161–166

(2015).

10. Yuyo, M. et al. A novel bone-thinning technique for transcranial stimulation motor-evoked potentials in rats. Sci. Rep. 11, 11

(2021).

11. Kjell, J. & Olson, L. Rat models of spinal cord injury: From pathology to potential therapies. Dis. Models Mech. 9, 1125–1137 (2016).

12. Iyer, S., Maybhate, A., Presacco, A. & All, A. H. Multi-limb acquisition of motor evoked potentials and its application in spinal

cord injury. J. Neurosci. Met. 193, 210–216 (2010).

13. Redondo-Castro, E., Navarro, X. & García-Alías, G. Longitudinal evaluation of residual cortical and subcortical motor evoked

potentials in spinal cord injured rats. J. Neurotrauma 33, 907–916 (2016).

14. Luft, A. R. et al. Transcranial magnetic stimulation in the rat. Exp. Brain Res. 140, 112–121 (2001).

15. Xie, C., Li, X., Fang, L. & Wang, T. Effects of athermal shortwave diathermy treatment on somatosensory evoked potentials and

motor evoked potentials in rats with spinal cord injury. Spine 44, E749–E758 (2019).

16. Xing, J., Katayama, Y., Yamamoto, T., Hirayama, T. & Tsubokawa, T. Quantitative evaluation of hemiparesis with corticomyographic

motor evoked potential recorded by transcranial magnetic stimulation. J. Neurotrauma 7, 57–64 (1990).

17. Tatsuya, S. et al. Avoidance of cerebral infarction in the territory of perforating arteries using intraoperative motor evoked potential

in aneurysm surgery: Current status and limitation. Surg. Cereb. Stroke 42, 347–352 (2014).

18. Tanaka, S., Tashiro, T., Gomi, A., Takanashi, J. & Ujjie, H. Sensitivity and specificity in transcranial motor-evoked potential monitoring during neurosurgical operations. Surg. Neurol. Int. 2, 111 (2011).

19. Murphy, P. G., Grondin, J., Altares, M. & Richardson, P. M. Induction of interleukin-6 in axotomized sensory neurons. J. Neurosci.

15, 5130–5138 (1995).

20. Gurcan, O. et al. Effect of asiatic acid on the treatment of spinal cord injury: An experimental study in rats. Turk. Neurosurg. 27,

259–264 (2017).

21. Imura, T. et al. Hypoxic preconditioning increases the neuroprotective effects of mesenchymal stem cells in a rat model of spinal

cord injury. J. Stem Cell Res. Ther. 7, 1 (2017).

22. Donnelly, D. J. & Popovich, P. G. Inflammation and its role in neuroprotection, axonal regeneration and functional recovery after

spinal cord injury. Exp. Neurol. 209, 378–388 (2008).

23. Sharma, H. S. et al. Topical application of TNF-alpha antiserum attenuates spinal cord trauma induced edema formation, microvascular permeability disturbances and cell injury in the rat. Acta Neurochir. Suppl. 86, 407–413 (2003).

24. Degterev, A. et al. Chemical inhibitor of nonapoptotic cell death with therapeutic potential for ischemic brain injury. Nat. Chem.

Biol. 1, 112–119 (2005).

25. Lu, J. V., Chen, H. C. & Walsh, C. M. Necroptotic signaling in adaptive and innate immunity. Semin. Cell Dev. Biol. 35, 33–39

(2014).

26. Fan, H. et al. Reactive astrocytes undergo M1 microglia/macrophages-induced necroptosis in spinal cord injury. Mol. Neurodegener.

11, 14 (2016).

27. Albayrak, Ö. et al. Mesenchymal stem cell therapy improves erectile dysfunction in experimental spinal cord injury. Int. J. Impot.

Res. 32, 308–316 (2020).

28. Wang, L. et al. Mesenchymal stem cell-derived exosomes reduce A1 astrocytes via downregulation of phosphorylated NFκB P65

subunit in spinal cord injury. Cell Physiol. Biochem. 50, 1535–1559 (2018).

29. Mothe, A. J. & Tator, C. H. Advances in stem cell therapy for spinal cord injury. J. Clin. Invest. 122, 3824–3834 (2012).

30. Orzalli, M. H. & Kagan, J. C. Apoptosis and necroptosis as host defense strategies to prevent viral infection. Trends Cell Biol. 27,

800–809 (2017).

31. Weinlich, R., Oberst, A., Beere, H. M. & Green, D. R. Necroptosis in development, inflammation and disease. Nat. Rev. Mol. Cell

Biol. 18, 127–136 (2017).

32. Hsu, H., Huang, J., Shu, H. B., Baichwal, V. & Goeddel, D. V. TNF-dependent recruitment of the protein kinase RIP to the TNF

receptor-1 signaling complex. Immunity 4, 387–396 (1996).

33. Thornton, C. & Hagberg, H. Role of mitochondria in apoptotic and necroptotic cell death in the developing brain. Clin. Chim.

Acta 7, 35–38 (2015).

34. Micheau, O. & Tschopp, J. Induction of TNF receptor I-mediated apoptosis via two sequential signaling complexes. Cell 114,

181–190 (2003).

35. Li, S. J. et al. The role of TNF-alpha, IL-6, IL-10, and GDNF in neuronal apoptosis in neonatal rat with hypoxic-ischemic encephalopathy. Eur. Rev. Med. Pharmacol. Sci. 18, 905–909 (2014).

36. Morrice, J. R., Gregory-Evans, C. Y. & Shaw, C. A. Necroptosis in amyotrophic lateral sclerosis and other neurological disorders.

Biochim. Biophys. Acta Mol. Basis Dis. 1863, 347–353 (2017).

37. Chen, S. D., Wu, C. L., Hwang, W. C. & Yang, D. I. More Insight into BDNF against neurodegeneration: Anti-apoptosis, antioxidation, and suppression of autophagy. Int. J. Mol. Sci. 18, 545 (2017).

38. Ying, X. et al. Hyperbaric oxygen therapy reduces apoptosis and dendritic/synaptic degeneration via the BDNF/TrkB signaling

pathways in SCI rats. Life Sci. 229, 187–199 (2019).

39. Jiang, Y. L., Liu, W. W., Wang, Y. & Yang, W. Y. MiR-210 suppresses neuronal apoptosis in rats with cerebral infarction through

regulating VEGF-notch signaling pathway. Eur. Rev. Med. Pharmacol. Sci. 24, 4971–4918 (2020).

40. Su, R., Su, W. & Jiao, Q. NGF protects neuroblastoma cells against beta-amyloid-induced apoptosis via the Nrf2/HO-1 pathway.

FEBS Open Bio 9, 2063–2071 (2019).

Scientific Reports |

Vol:.(1234567890)

(2021) 11:21907 |

https://doi.org/10.1038/s41598-021-01490-1

10

www.nature.com/scientificreports/

41. Montero, M. L. et al. Docosahexaenoic acid protection against palmitic acid-induced lipotoxicity in NGF-differentiated PC12 cells

involves enhancement of autophagy and inhibition of apoptosis and necroptosis. J. Neurochem. 155, 559–576 (2020).

42. Saeidikhoo, S. et al. Effect of Sertoli cell transplantation on reducing neuroinflammation-induced necroptosis and improving

motor coordination in the rat model of cerebellar ataxia induced by 3-acetylpyridine. J. Mol. Neurosci. 70, 1153–1163 (2020).

43. Otsuka, T. et al. Comparisons of neurotrophic effects of mesenchymal stem cells derived from different tissue on chronic spinal

cord injury rats. Stem Cells Dev. 30, 865–875 (2021).

44. Pomies, P. et al. Involvement of the FoxO1/MuRF1/Atrogin-1 signaling pathway in the oxidative stress-induced atrophy of cultured

chronic obstructive pulmonary disease myotubes. PLoS ONE 11, e0160092 (2016).

45. Gutierrez-Fernandez, M. et al. Effects of intravenous administration of allogenic bone marrow- and adipose tissue-derived mesenchymal stem cells on functional recovery and brain repair markers in experimental ischemic stroke. Stem Cell Res. Ther. 4, 11

(2013).

46. Mitsuhara, T. et al. Simulated microgravity facilitates cell migration and neuroprotection after bone marrow stromal cell transplantation in spinal cord injury. Stem Cell Res. Ther. 4, 35 (2013).

47. Basso, D. M., Beattie, M. S. & Bresnahan, J. C. Graded histological and locomotor outcomes after spinal cord contusion using the

NYU weight-drop device versus transection. Exp. Neurol. 139, 244–256 (1996).

48. Sakai, K. et al. Human dental pulp-derived stem cells promote locomotor recovery after complete transection of the rat spinal cord

by multiple neuro-regenerative mechanisms. J. Clin. Invest. 122, 80–90 (2012).

49. Basso, D. M., Beattie, M. S. & Bresnahan, J. C. A sensitive and reliable locomotor rating scale for open field testing in rats. J. Neurotrauma 12, 1–21 (1995).

50. Wells, J. E. et al. An adverse role for matrix metalloproteinase 12 after spinal cord injury in mice. J. Neurosci. 23, 10107–10115

(2003).

51. Rivlin, A. S. & Tator, C. H. Objective clinical assessment of motor function after experimental spinal cord injury in the rat. J.

Neurosurg. 47, 577–581 (1977).

52. Sekine, Y. et al. The Kelch repeat protein KLHDC10 regulates oxidative stress-induced ASK1 activation by suppressing PP5. Mol.

Cell. 48, 692–704 (2012).

53. Neirinckx, V. et al. Adult bone marrow mesenchymal and neural crest stem cells are chemoattractive and accelerate motor recovery

in a mouse model of spinal cord injury. Stem Cell Res. Ther. 6, 211 (2015).

Author contributions

Y.M. and M.T. recorded tcMEP. Histological analysis and mRNA expression analysis were performed by Y.M,

T.O, and L.Y. Animal experimentation was performed by Y.M, T.M, T.O, M.T, K.S, M.K, and M.H. Y.M, T.M,

T.O, and M.T. performed all methodologies. Funding was acquired by T.M. Y.M wrote the initial version of the

manuscript. All authors contributed to and approved the final version of the manuscript.

Funding

This work was supported in part by a Grant-in-Aid for Scientific Research from the Japan Society for the Promotion of Science (JSPS KAKENHI Grant Number 18K16561) and TWO CELLS Co., Ltd., Hiroshima, Japan.

Competing interests The authors declare no competing interests.

Additional information

Supplementary Information The online version contains supplementary material available at https://​doi.​org/​

10.​1038/​s41598-​021-​01490-1.

Correspondence and requests for materials should be addressed to Y.M.

Reprints and permissions information is available at www.nature.com/reprints.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and

institutional affiliations.

Open Access This article is licensed under a Creative Commons Attribution 4.0 International

License, which permits use, sharing, adaptation, distribution and reproduction in any medium or

format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the

Creative Commons licence, and indicate if changes were made. The images or other third party material in this

article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the

material. If material is not included in the article’s Creative Commons licence and your intended use is not

permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from

the copyright holder. To view a copy of this licence, visit http://​creat​iveco​mmons.​org/​licen​ses/​by/4.​0/.

© The Author(s) 2021

Scientific Reports |

(2021) 11:21907 |

https://doi.org/10.1038/s41598-021-01490-1

11

Vol.:(0123456789)

...

参考文献をもっと見る