Korean J Pain.  2014 Jul;27(3):239-245. 10.3344/kjp.2014.27.3.239.

Intrathecal Administration of Mesenchymal Stem Cells Reduces the Reactive Oxygen Species and Pain Behavior in Neuropathic Rats

Affiliations
  • 1Department of Anesthesiology and Pain Medicine, School of Medicine, Chungnam National University, Daejeon, Korea. whlee@cnu.ac.kr
  • 2Department of Microbiology, School of Medicine, Chungnam National University, Daejeon, Korea. songch@cnu.ac.kr

Abstract

BACKGROUND
Neuropathic pain induced by spinal or peripheral nerve injury is very resistant to common pain killers, nerve block, and other pain management approaches. Recently, several studies using stem cells suggested a new way to control the neuropatic pain. In this study, we used the spinal nerve L5 ligation (SNL) model to investigate whether intrathecal rat mesenchymal stem cells (rMSCs) were able to decrease pain behavior, as well as the relationship between rMSCs and reactive oxygen species (ROS).
METHODS
Neuropathic pain of the left hind paw was induced by unilateral SNL in Sprague-Dawley rats (n = 10 in each group). Mechanical sensitivity was assessed using Von Frey filaments at 3, 7, 10, 12, 14, 17, and 24 days post-ligation. rMSCs (10 microl, 1 x 105) or phosphate buffer saline (PBS, 10 microl) was injected intrathecally at 7 days post-ligation. Dihydroethidium (DHE), an oxidative fluorescent dye, was used to detect ROS at 24 days post-ligation.
RESULTS
Tight ligation of the L5 spinal nerve induced allodynia in the left hind paw after 3 days post-ligation. ROS expression was increased significantly (P < 0.05) in spinal dorsal horn of L5. Intrathecal rMSCs significantly (P < 0.01) alleviated the allodynia at 10 days after intrathecal injection (17 days post-ligation). Intrathecal rMSCs administration significantly (P < 0.05) reduced ROS expression in the spinal dorsal horn.
CONCLUSIONS
These results suggest that rMSCs may modulate neuropathic pain generation through ROS expression after spinal nerve ligation.

Keyword

mesenchymal stem cells; neuropathic pain; reactive oxygen species

MeSH Terms

Animals
Horns
Hyperalgesia
Injections, Spinal
Ligation
Mesenchymal Stromal Cells*
Nerve Block
Neuralgia
Pain Management
Peripheral Nerve Injuries
Rats*
Rats, Sprague-Dawley
Reactive Oxygen Species*
Spinal Nerves
Stem Cells
Reactive Oxygen Species

Figure

  • Fig. 1 Von Frey filament testing results. (A) Effect of spinal nerve ligation on the development of neuropathic pain. A significant decrease in the paw withdrawal threshold in the Von Frey filament test is induced 3 days after SNL. ***P < 0.001 vs. before surgery. (B) Effect of rMSCs on paw withdrawal reflex responses to Von Frey filament stimuli in rat pain models. rMSCs treatment on day 7 after SNL significantly increased the withdrawal threshold on days 10 and 17 after administration. **P < 0.01 vs. PBS group and #P < 0.05, ##P < 0.01 vs. Pain group. Values are expressed as the mean ± SEM (n = 10 in each group) Abbreviations: T, post-administration.

  • Fig. 2 DHE staining of the L5 spinal dorsal horn on day 24 following the spinal nerve ligation. (A) The L5 spinal dorsal horn segments (naive, pain, PBS, rMSCs groups) were stained with ROS-sensitive dye DHE (dihydroethidium). ROS (red) expression was decreased in the spinal cord of the rMSCs group (scale bar, 200 µm). (B) Fluorescent intensities of DHE signals were measured and compared with other groups. The intensities of DHE in the pain and PBS groups show a significant difference at *P < 0.05 compared with the naïve rats. The rMSCs group shows a statistical difference at #P < 0.05 compared with the pain or PBS groups. Values are expressed as the mean ± SEM.


Cited by  3 articles

Stem cell therapy in pain medicine
Yong Hee Han, Kyung Hoon Kim, Salahadin Abdi, Tae Kyun Kim
Korean J Pain. 2019;32(4):245-255.    doi: 10.3344/kjp.2019.32.4.245.

Antinociceptive and neuroprotective effects of bromelain in chronic constriction injury-induced neuropathic pain in Wistar rats
Ahmed Olalekan Bakare, Bamidele Victor Owoyele
Korean J Pain. 2020;33(1):13-22.    doi: 10.3344/kjp.2020.33.1.13.

The effect of human mesenchymal stem cell injection on pain behavior in chronic post-ischemia pain mice
Sie Hyeon Yoo, Sung Hyun Lee, Seunghwan Lee, Jae Hong Park, Seunghyeon Lee, Heecheol Jin, Hue Jung Park
Korean J Pain. 2020;33(1):23-29.    doi: 10.3344/kjp.2020.33.1.23.


Reference

1. Gilron I, Watson CP, Cahill CM, Moulin DE. Neuropathic pain: a practical guide for the clinician. CMAJ. 2006; 175:265–275. PMID: 16880448.
Article
2. Jaiswal RK, Jaiswal N, Bruder SP, Mbalaviele G, Marshak DR, Pittenger MF. Adult human mesenchymal stem cell differentiation to the osteogenic or adipogenic lineage is regulated by mitogen-activated protein kinase. J Biol Chem. 2000; 275:9645–9652. PMID: 10734116.
Article
3. Conget PA, Minguell JJ. Phenotypical and functional properties of human bone marrow mesenchymal progenitor cells. J Cell Physiol. 1999; 181:67–73. PMID: 10457354.
Article
4. Lu L, Zhao C, Liu Y, Sun X, Duan C, Ji M, et al. Therapeutic benefit of TH-engineered mesenchymal stem cells for Parkinson's disease. Brain Res Brain Res Protoc. 2005; 15:46–51. PMID: 15878150.
Article
5. Yang LY, Huang TH, Ma L. Bone marrow stromal cells express neural phenotypes in vitro and migrate in brain after transplantation in vivo. Biomed Environ Sci. 2006; 19:329–335. PMID: 17190183.
6. Le Blanc K, Pittenger M. Mesenchymal stem cells: progress toward promise. Cytotherapy. 2005; 7:36–45. PMID: 16040382.
Article
7. Kim HK, Park SK, Zhou JL, Taglialatela G, Chung K, Coggeshall RE, et al. Reactive oxygen species (ROS) play an important role in a rat model of neuropathic pain. Pain. 2004; 111:116–124. PMID: 15327815.
Article
8. Wang ZQ, Porreca F, Cuzzocrea S, Galen K, Lightfoot R, Masini E, et al. A newly identified role for superoxide in inflammatory pain. J Pharmacol Exp Ther. 2004; 309:869–878. PMID: 14988418.
Article
9. Valle-Prieto A, Conget PA. Human mesenchymal stem cells efficiently manage oxidative stress. Stem Cells Dev. 2010; 19:1885–1893. PMID: 20380515.
Article
10. Tal M, Bennett GJ. Extra-territorial pain in rats with a peripheral mononeuropathy: mechano-hyperalgesia and mechano-allodynia in the territory of an uninjured nerve. Pain. 1994; 57:375–382. PMID: 7936715.
Article
11. Soleimani M, Nadri S. A protocol for isolation and culture of mesenchymal stem cells from mouse bone marrow. Nat Protoc. 2009; 4:102–106. PMID: 19131962.
Article
12. Yang CC, Shih YH, Ko MH, Hsu SY, Cheng H, Fu YS. Transplantation of human umbilical mesenchymal stem cells from Wharton's jelly after complete transection of the rat spinal cord. PLoS One. 2008; 3:e3336. PMID: 18852872.
Article
13. Papir-Kricheli D, Frey J, Laufer R, Gilon C, Chorev M, Selinger Z, et al. Behavioural effects of receptor-specific substance P agonists. Pain. 1987; 31:263–276. PMID: 2448728.
Article
14. Harraz MM, Marden JJ, Zhou W, Zhang Y, Williams A, Sharov VS, et al. SOD1 mutations disrupt redox-sensitive Rac regulation of NADPH oxidase in a familial ALS model. J Clin Invest. 2008; 118:659–670. PMID: 18219391.
Article
15. Kumar S, Ruchi R, James SR, Chidiac EJ. Gene therapy for chronic neuropathic pain: how does it work and where do we stand today? Pain Med. 2011; 12:808–822. PMID: 21564510.
Article
16. Marchand F, Perretti M, McMahon SB. Role of the immune system in chronic pain. Nat Rev Neurosci. 2005; 6:521–532. PMID: 15995723.
Article
17. Hill RG. Molecular basis for the perception of pain. Neuroscientist. 2001; 7:282–292. PMID: 11488394.
18. Scholz J, Woolf CJ. The neuropathic pain triad: neurons, immune cells and glia. Nat Neurosci. 2007; 10:1361–1368. PMID: 17965656.
Article
19. Reichling DB, Levine JD. Critical role of nociceptor plasticity in chronic pain. Trends Neurosci. 2009; 32:611–618. PMID: 19781793.
Article
20. Kim D, You B, Jo EK, Han SK, Simon MI, Lee SJ. NADPH oxidase 2-derived reactive oxygen species in spinal cord microglia contribute to peripheral nerve injury-induced neuropathic pain. Proc Natl Acad Sci U S A. 2010; 107:14851–14856. PMID: 20679217.
Article
21. Siniscalco D, Giordano C, Rossi F, Maione S, de Novellis V. Role of neurotrophins in neuropathic pain. Curr Neuropharmacol. 2011; 9:523–529. PMID: 22654713.
Article
22. Wagers AJ, Weissman IL. Plasticity of adult stem cells. Cell. 2004; 116:639–648. PMID: 15006347.
Article
23. Siniscalco D, Giordano C, Galderisi U, Luongo L, Alessio N, Di Bernardo G, et al. Intra-brain microinjection of human mesenchymal stem cells decreases allodynia in neuropathic mice. Cell Mol Life Sci. 2010; 67:655–669. PMID: 19937263.
Article
24. Savitz SI, Dinsmore JH, Wechsler LR, Rosenbaum DM, Caplan LR. Cell therapy for stroke. NeuroRx. 2004; 1:406–414. PMID: 15717044.
Article
25. Levy YS, Bahat-Stroomza M, Barzilay R, Burshtein A, Bulvik S, Barhum Y, et al. Regenerative effect of neural-induced human mesenchymal stromal cells in rat models of Parkinson's disease. Cytotherapy. 2008; 10:340–352. PMID: 18574767.
Article
26. Zurita M, Vaquero J. Functional recovery in chronic paraplegia after bone marrow stromal cells transplantation. Neuroreport. 2004; 15:1105–1108. PMID: 15129154.
Article
27. Kim CH, Kim YW, Jang SH, Chang CH, Jung JH, Kim SH. Motor function recovery after adipose tissue derived mesenchymal stem cell therapy in rats with cerebral infarction. J Korean Neurosurg Soc. 2006; 40:267–272.
28. Park ES, Gao X, Chung JM, Chung K. Levels of mitochondrial reactive oxygen species increase in rat neuropathic spinal dorsal horn neurons. Neurosci Lett. 2006; 391:108–111. PMID: 16183198.
Article
Full Text Links
  • KJP
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