J Vet Sci.  2017 Sep;18(3):377-386. 10.4142/jvs.2017.18.3.377.

Effect of canine mesenchymal stromal cells overexpressing heme oxygenase-1 in spinal cord injury

Affiliations
  • 1BK21 PLUS Program for Creative Veterinary Science Research, Research Institute for Veterinary Science and College of Veterinary Medicine, Seoul National University, Seoul 08872, Korea. ohkweon@snu.ac.kr

Abstract

Heme oxygenase-1 (HO-1) is a stress-responsive enzyme that modulates the immune response and oxidative stress associated with spinal cord injury (SCI). This study aimed to investigate neuronal regeneration via transplantation of mesenchymal stromal cells (MSCs) overexpressing HO-1. Canine MSCs overexpressing HO-1 were generated by using a lentivirus packaging protocol. Eight beagle dogs with experimentally-induced SCI were divided into GFP-labeled MSC (MSC-GFP) and HO-1-overexpressing MSC (MSC-HO-1) groups. MSCs (1 × 10⁷ cells) were transplanted at 1 week after SCI. Spinal cords were harvested 8 weeks after transplantation, after which histopathological, immunofluorescence, and western blot analyses were performed. The MSC-HO-1 group showed significantly improved functional recovery at 7 weeks after transplantation. Histopathological results showed fibrotic changes and microglial cell infiltration were significantly decreased in the MSC-HO-1 group. Immunohistochemical (IHC) results showed significantly increased expression levels of HO-1 and neuronal markers in the MSC-HO-1 group. Western blot results showed significantly decreased expression of tumor necrosis factor-alpha, interleukin-6, cycloogygenase 2, phosphorylated-signal transducer and activator of transcription 3, and galactosylceramidase in the MSC-HO-1 group, while expression levels of glial fibrillary acidic protein, β3-tubulin, neurofilament medium, and neuronal nuclear antigen were similar to those observed in IHC results. Our results demonstrate that functional recovery after SCI can be promoted to a greater extent by transplantation of HO-1-overexpressing MSCs than by normal MSCs.

Keyword

heme oxygenase-1; mesenchymal stromal cells; spinal cord injuries

MeSH Terms

Animals
Blotting, Western/veterinary
Dogs/*injuries/metabolism
Female
Heme Oxygenase-1/*metabolism
Mesenchymal Stromal Cells/*enzymology/metabolism
Spinal Cord/metabolism/pathology
Spinal Cord Injuries/metabolism/pathology/*veterinary
Heme Oxygenase-1

Figure

  • Fig. 1 Fluorescence microscopy images and cell viability results. (A) Green fluorescence protein (GFP) overexpressing cADMSCs. Top panels, cADMSC; middle panels, GFP-labeled cADMSCs (MSC-GFP); lower panels, GFP-labeled HO-1 overexpressing cADMSCs (MSC-HO-1). GFP expression was identified in MSC-GFP and MSC-HO-1 via fluorescence microscopy. Right panels are higher magnification images of left panels, respectively. (B) Cell viability of the three cell types showing no significant difference. Scale bars = 50 µm.

  • Fig. 2 Protein expression in cADMSCs overexpressing heme oxygenase 1 (HO-1) and their antioxidant effects. (A) Western blot results for cADMSCs, MSC-GFP, and MSC-HO-1. Expression of HO-1 in MSC-HO-1 was significantly higher than it was in the cADMSCs and MSC-GFP, and the MSC-GFP and MSC-HO-1 showed a significant increase in GFP expression relative to that in cADMSCs, but there was no difference between overexpressed cells (*p < 0.05 vs. cADMSCs, †p < 0.05 vs. MSC-GFP). (B) Antioxidant capacity of cADMSCs, MSC-GFP, and MSC-HO-1. Uric acid equivalent level of MSC-HO-1 was significantly higher than that of the other groups. There was no difference between the cADMSCs and MSC-GFP (*p < 0.05 vs. PBS, †p < 0.05 vs. cADMSCs, and ‡p < 0.05 vs. MSC-GFP).

  • Fig. 3 Behavioral analysis results using Basso, Beattie, and Bresnahan (BBB) scores and Tarlov scales. (A) BBB scores before and during the 8 weeks after transplantation. Post-transplantation, the BBB scores of the MSC-HO-1 group were significantly improved at 7 and 8 weeks after transplantation compared with the GFP-labeled cADMSCs (MSC-GFP) scores (*p < 0.05). Hindlimb locomotion at 8 weeks after transplantation were determined by two grading system: (B) revised Tarlov scales and (C) modified Tarlov scales. Higher motor function recovery was indicated in the MSC-HO-1 group than the MSC-GFP group (*p < 0.05).

  • Fig. 4 Histopathological analysis with H&E staining. Histological analysis of spinal cord lesions stained with H&E. (A) MSC-GFP group; upon magnification of the injury epicenter, fibroblast-like cell proliferation was detected. (B) MSC-HO-1 group; spinal cord lesion was limited to the compression region and a greater reduction in fibrotic changes was observed compared with those in the MSC-GFP group. Also, upon magnification, a reduction in fibroblast-like cell proliferation was observed. (C) Quantification of fibrotic tissue was showed significant difference between groups (*p < 0.05). 40× (top panels of A and B), 200× (bottom panels of A and B; enlargements of squares of the middle panels).

  • Fig. 5 Immunohistochemical assessments. (A) Immunofluorescence staining at 8 weeks after transplantation. Injured spinal cord lesions were immunostained for heme oxygenase 1 (HO-1), glial fibrillary acidic protein (GFAP), β3-tubulin, neurofilament M (NF-M), neuronal nuclear antigen (NeuN), galactosylceramidase (GALC), and phosphorylated-signal transducer and activator of transcription 3 (p-STAT3; red); transplanted cells (green fluorescent protein [GFP]); each nucleus was stained with DAPI (blue). (B), (C), and (D) showed quantification of immunostaining results. (B) Expression of red positive markers of antibodies. Expressions of the HO-1, β3-tubulin, NF-M, and NeuN were higher in the MSC-HO-1 group than MSC-GFP group, and expression of GFAP was adversely affected. The number of transplanted cells expressing GFP was not different between the two groups (*p < 0.05). (C) GFP expression shows no difference between the groups. (D) Comparison of the origins of HO-1 and neural cells in the injured site, whether from implanted MSCs or endogenous host cells. The expression of most neural cell markers from endogenous origins (not GFP labeled) were significantly higher than those of transplanted cells origins (GFP labeled) in both groups (*p < 0.05). However, expression rate of HO-1 marker from transplanted cells origins was significantly higher than that from endogenous origin cells in the MSC-HO-1 group (*p < 0.05). Scale bars = 50 µm.

  • Fig. 6 Western blot analysis. Protein expressions at 8 weeks after transplantation were evaluated by western blotting. Overexpression markers, heme oxygenase 1 (HO-1) and green fluorescent protein (GFP); inflammatory markers, tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), cycloogygenase 2 (COX2), phosphorylated-signal transducer and activator of transcription 3 (p-STAT3), and GALC; neuronal markers, glial fibrillary acidic protein (GFAP), β3-tubulin, NF-M, and neuronal nuclear antigen (NeuN). Data presented as means ± SE of three independent experiments. The graph and bars depict means ± SE of four dogs per group, as determined by observed densitometry relative to β-actin (*p < 0.05, †p < 0.001).


Reference

1. Andrews EM, Tsai SY, Johnson SC, Farrer JR, Wagner JP, Kopen GC, Kartje GL. Human adult bone marrow-derived somatic cell therapy results in functional recovery and axonal plasticity following stroke in the rat. Exp Neurol. 2008; 211:588–592.
Article
2. Bains M, Hall ED. Antioxidant therapies in traumatic brain and spinal cord injury. Biochim Biophys Acta. 2012; 1822:675–684.
Article
3. Barros Filho TE, Molina AE. Analysis of the sensitivity and reproducibility of the Basso, Beattie, Bresnahan (BBB) scale in Wistar rats. Clinics (Sao Paulo). 2008; 63:103–108.
Article
4. Bradford MM. A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem. 1976; 72:248–254.
Article
5. Cho SR, Kim YR, Kang HS, Yim SH, Park CI, Min YH, Lee BH, Shin JC, Lim JB. Functional recovery after the transplantation of neurally differentiated mesenchymal stem cells derived from bone barrow in a rat model of spinal cord injury. Cell Transplant. 2009; 18:1359–1368.
Article
6. Dougherty KD, Dreyfus CF, Black IB. Brain-derived neurotrophic factor in astrocytes, oligodendrocytes, and microglia/macrophages after spinal cord injury. Neurobiol Dis. 2000; 7:574–585.
Article
7. Emgård M, Hallin U, Karlsson J, Bahr BA, Brundin P, Blomgren K. Both apoptosis and necrosis occur early after intracerebral grafting of ventral mesencephalic tissue: a role for protease activation. J Neurochem. 2003; 86:1223–1232.
Article
8. Herrmann JE, Imura T, Song B, Qi J, Ao Y, Nguyen TK, Korsak RA, Takeda K, Akira S, Sofroniew MV. STAT3 is a critical regulator of astrogliosis and scar formation after spinal cord injury. J Neurosci. 2008; 28:7231–7243.
Article
9. Kanno H, Ozawa H, Dohi Y, Sekiguchi A, Igarashi K, Itoi E. Genetic ablation of transcription repressor Bach1 reduces neural tissue damage and improves locomotor function after spinal cord injury in mice. J Neurotrauma. 2009; 26:31–39.
Article
10. Keirstead HS, Nistor G, Bernal G, Totoiu M, Cloutier F, Sharp K, Steward O. Human embryonic stem cell-derived oligodendrocyte progenitor cell transplants remyelinate and restore locomotion after spinal cord injury. J Neurosci. 2005; 25:4694–4705.
Article
11. Kim M, Kim Y, Lee S, Kuk M, Kim AY, Kim W, Kweon OK. Comparison of viability and antioxidant capacity between canine adipose-derived mesenchymal stem cells and heme oxygenase-1-overexpressed cells after freeze-thawing. J Vet Med Sci. 2016; 78:619–625.
Article
12. Kim Y, Jo SH, Kim WH, Kweon OK. Antioxidant and anti-inflammatory effects of intravenously injected adipose derived mesenchymal stem cells in dogs with acute spinal cord injury. Stem Cell Res Ther. 2015; 6:229.
Article
13. Kuk M, Kim Y, Lee SH, Kim WH, Kweon OK. Osteogenic ability of canine adipose-derived mesenchymal stromal cell sheets in relation to culture time. Cell Transplant. 2016; 25:1415–1422.
Article
14. Lee SH, Kim Y, Rhew D, Kuk M, Kim M, Kim WH, Kweon OK. Effect of the combination of mesenchymal stromal cells and chondroitinase ABC on chronic spinal cord injury. Cytotherapy. 2015; 17:1374–1383.
Article
15. Lim JH, Byeon YE, Ryu HH, Jeong YH, Lee YW, Kim WH, Kang KS, Kweon OK. Transplantation of canine umbilical cord blood-derived mesenchymal stem cells in experimentally induced spinal cord injured dogs. J Vet Sci. 2007; 8:275–282.
Article
16. Lu P, Jones LL, Tuszynski MH. Axon regeneration through scars and into sites of chronic spinal cord injury. Exp Neurol. 2007; 203:8–21.
Article
17. Lu P, Jones LL, Tuszynski MH. BDNF-expressing marrow stromal cells support extensive axonal growth at sites of spinal cord injury. Exp Neurol. 2005; 191:344–360.
Article
18. Mothe AJ, Tator CH. Review of transplantation of neural stem/progenitor cells for spinal cord injury. Int J Dev Neurosci. 2013; 31:701–713.
Article
19. Öllinger R, Pratschke J. Role of heme oxygenase-1 in transplantation. Transpl Int. 2010; 23:1071–1081.
Article
20. Origassa CS, Câmara NO. Cytoprotective role of heme oxygenase-1 and heme degradation derived end products in liver injury. World J Hepatol. 2013; 5:541–549.
Article
21. Osaka M, Honmou O, Murakami T, Nonaka T, Houkin K, Hamada H, Kocsis JD. Intravenous administration of mesenchymal stem cells derived from bone marrow after contusive spinal cord injury improves functional outcome. Brain Res. 2010; 1343:226–235.
Article
22. Park SS, Byeon YE, Ryu HH, Kang BJ, Kim Y, Kim WH, Kang KS, Han HJ, Kweon OK. Comparison of canine umbilical cord blood-derived mesenchymal stem cell transplantation times: involvement of astrogliosis, inflammation, intracellular actin cytoskeleton pathways, and neurotrophin-3. Cell Transplant. 2011; 20:1867–1880.
Article
23. Park SS, Lee YJ, Lee SH, Lee D, Choi K, Kim WH, Kweon OK, Han HJ. Functional recovery after spinal cord injury in dogs treated with a combination of Matrigel and neural-induced adipose-derived mesenchymal stem cells. Cytotherapy. 2012; 14:584–597.
Article
24. Rabinowitz RS, Eck JC, Harper CM Jr, Larson DR, Jimenez MA, Parisi JE, Friedman JA, Yaszemski MJ, Currier BL. Urgent surgical decompression compared to methylprednisolone for the treatment of acute spinal cord injury: a randomized prospective study in beagle dogs. Spine (Phila Pa 1976). 2008; 33:2260–2268.
Article
25. Ryu HH, Byeon YE, Park SS, Kang BJ, Seo MS, Park SB, Kim WH, Kang KS, Kweon OK. Immunohistomorphometric analysis of transplanted umbilical cord blood-derived mesenchymal stem cells and rhe resulting anti-inflammatory effects on nerve regeneration of injured canine spinal cord. Tissue Eng Regen Med. 2011; 8:173–182.
26. Ryu HH, Lim JH, Byeon YE, Park JR, Seo MS, Lee YW, Kim WH, Kang KS, Kweon OK. Functional recovery and neural differentiation after transplantation of allogenic adipose-derived stem cells in a canine model of acute spinal cord injury. J Vet Sci. 2009; 10:273–284.
Article
27. Snyder EY, Teng YD. Stem cells and spinal cord repair. N Engl J Med. 2012; 366:1940–1942.
Article
28. Soares MP, Marguti I, Cunha A, Larsen R. Immunoregulatory effects of HO-1: how does it work? Curr Opin Pharmacol. 2009; 9:482–489.
Article
29. Tang YL, Tang Y, Zhang YC, Qian K, Shen L, Phillips MI. Improved graft mesenchymal stem cell survival in ischemic heart with a hypoxia-regulated heme oxygenase-1 vector. J Am Coll Cardiol. 2005; 46:1339–1350.
Article
30. Taylor L, Jones L, Tuszynski MH, Blesch A. Neurotrophin-3 gradients established by lentiviral gene delivery promote short-distance axonal bridging beyond cellular grafts in the injured spinal cord. J Neurosci. 2006; 26:9713–9721.
Article
31. Torres-Espín A, Redondo-Castro E, Hernandez J, Navarro X. Immunosuppression of allogenic mesenchymal stem cells transplantation after spinal cord injury improves graft survival and beneficial outcomes. J Neurotrauma. 2015; 32:367–380.
Article
32. Uemura M, Refaat MM, Shinoyama M, Hayashi H, Hashimoto N, Takahashi J. Matrigel supports survival and neuronal differentiation of grafted embryonic stem cell-derived neural precursor cells. J Neurosci Res. 2010; 88:542–551.
Article
33. Yamauchi T, Lin Y, Sharp FR, Noble-Haeusslein LJ. Hemin induces heme oxygenase-1 in spinal cord vasculature and attenuates barrier disruption and neutrophil infiltration in the injured murine spinal cord. J Neurotrauma. 2004; 21:1017–1030.
Article
34. Yeom HJ, Koo OJ, Yang J, Cho B, Hwang JI, Park SJ, Hurh S, Kim H, Lee EM, Ro H, Kang JT, Kim SJ, Won JK, O'Connell PJ, Kim H, Surh CD, Lee BC, Ahn C. Generation and characterization of human heme oxygenase-1 transgenic pigs. PLoS One. 2012; 7:e46646.
Article
35. Zhang LF, Qi J, Zuo G, Jia P, Shen X, Shao J, Kang H, Yang H, Deng L. Osteoblast-secreted factors promote proliferation and osteogenic differentiation of bone marrow stromal cells via VEGF/heme-oxygenase-1 pathway. PLoS One. 2014; 9:e99946.
Article
36. Zhang M, Zhang BH, Chen L, An W. Overexpression of heme oxygenase-1 protects smooth muscle cells against oxidative injury and inhibits cell proliferation. Cell Res. 2002; 12:123–132.
Article
Full Text Links
  • JVS
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