The efficacy of cryopreserved ex vivo expanded rat bone marrow-derived multipotent mesenchymal stromal cells in the repair of radiation injuries in rats

Home/2022, Vol. 10, No. 1/The efficacy of cryopreserved ex vivo expanded rat bone marrow-derived multipotent mesenchymal stromal cells in the repair of radiation injuries in rats

Cell and Organ Transplantology. 2022; 10(1):10-16.
DOI: 10.22494/cot.v10i1.139

The efficacy of cryopreserved ex vivo expanded rat bone marrow-derived multipotent mesenchymal stromal cells in the repair of radiation injuries in rats

Uzlenkova N., Skorobogatova N., Kryvko A., Krasnoselsky M.

  • Grigoriev Institute for medical Radiology of the National Academy of Medical Sciences of Ukraine, Kharkiv, Ukraine

Abstract

At present, applying multipotent mesenchymal stromal cells (MSCs) as cell therapy for radiation damages have gained increasing attention since current medical management remains far from satisfactory.
The aim of the study is to examine the efficacy of cryopreserved ex vivo expanded bone marrow-derived MSCs (rBM-MSCs) to the repair of radiation injuries on rat models of total and local radiation exposure.
Materials and methods. The MSCs were derived from bone marrow of non-irradiated female albino rats aged 4 months, short-term ex vivo expanded for two passages and cryopreserved under dimethyl sulfoxide cryoprotection for low temperature storage at -70 oC for 6-12 months. The cryopreserved samples from each batch of rBM-MSCs culture were tested for the viability and functional characteristics before being transplanted to rats in experiments in vivo. The acute radiation damages in rats were modeled by total body irradiation (TBI) at doses of 5.5 Gy (TBI 5.5) and 7.0 Gy (TBI 7.0) and locally irradiated in the right hip skin at a dose of 50 Gy. The cryopreserved rBM-MSCs (1.5•106 and 0.5•106 cells/animal) were intravenously transplanted within 24 h following TBI and locally injected (twice 1.5•106 Cells/animals) on days 15 and 21 following thigh irradiation. The efficacy of cryopreserved rBM-MSCs was assessed by survival and hematological study as well as the irradiated skin wound healing assay.
Results. The cryopreserved ex vivo expanded rBM-MSCs were characterized by high level of functional activity with cell viability about 80 %, include at least 8.5 % of the colony forming MSCs and MSCs with ability to adipogenic and osteogenic differentiation In TBI 5.5 rats, cryopreserved transplanted rBM-MSCs (1.5·106 cells/animal) prevented acute leukopenia in the first critical days of the radiation injury by increasing the number of leukocytes by 3.7 times on day 2 and contributed to a more complete recovery of hematological disorders by increasing the BM cells number and platelet count on day 22, which led to the increase of the increase of overall survival up to 100 % with a regain of body weight. In TBI 7.0 rats, the lower transplanted dose of rBM-MSCs (0.5•106 cells/animal) was more effective in terms of general recovery and extended the overall survival time for 6 days. The locally injected rBM-MSCs (twice 1.5•106 cells/animals) reduced the severity and promoted the healing of radiation skin wounds according to the results of scoring and wound size assay.
Conclusion. The present study confirms that the cryopreserved ex vivo expanded rBM-MSCs were functionally complete for the therapeutic use on rat models of experimental radiation damage and were effective for the recovery of hematopoietic system and severe skin wound after radiation exposure.

Key words: bone marrow-derived multipotent mesenchymal stromal cells; cell expansion ex vivo; cell cryopreservation; radiation injury

Full Text PDF

1. McBride WH, Schaue D. Radiation‐induced tissue damage and response. J Pathol. 2020; 250 (5):647-655.https://doi 10.1002/path.5389
https://doi.org/10.1002/path.5389
PMid:31990369 PMCid:PMC7216989
2. Hofer M, Pospisil M, Komurkova D, Hoferova Z. Granulocyte colony‐stimulating factor in the treatment of acute radiation syndrome: a concise review. Molecules. 2014;19:4770-8. https://doi: 10.3390/molecules19044770.
https://doi.org/10.3390/molecules19044770
PMid:24743934 PMCid:PMC6270858
3. MacVittie TJ, Farese AM, Jackson W 3rd. Defining the full therapeutic potential of recombinant growth factors in the post radiation‐accident environment: the effect of supportive care plus administration of G‐CSF. Health Phys. 2005;89:546-555. https://doi: 10.1097/01.hp.0000173143.69659.5b.
https://doi.org/10.1097/01.HP.0000173143.69659.5b
PMid:16217198
4. DiCarlo AL, et al. Cellular therapies for treatment of radiation injury: report from a NIH/NIAID and IRSN workshop. Radiat. Res. 2017; 188:54-75.https://doi: 10.1667/RR14810.1. Epub 2017 Jun 12.
https://doi.org/10.1667/RR14810.1
PMid:28605260 PMCid:PMC5564392
5. Rios C, Jourdain J-R, DiCarlo AL. Cellular therapies for treatment of radiation injury after a mass casualty incident. Radiat. Res. 2017
https://doi.org/10.1667/RR14835.1
PMid:28609636 PMCid:PMC5564290
188 (2):242-5. https://doi: 10.1667/RR14835.1. Epub 2017 Jun 13.
https://doi.org/10.1667/RR14835.1
PMid:28609636 PMCid:PMC5564290
6. Pittenger MF, et al. Mesenchymal stem cell perspective: cell biology to clinical progress. NPJ Regen. Med. 2019;4:22-36. https://doi: 10.1038/s41536-019-0083-6. eCollection 2019.
https://doi.org/10.1038/s41536-019-0083-6
PMid:31815001 PMCid:PMC6889290
7. Qi K, Li N, Zhang Z, Melino G. Tissue regeneration: The crosstalk between mesenchymal stem cells and immune response. Cell Immunol. 2018;326:86-93. https://doi: 10.1016/j.cellimm.2017.11.010.
https://doi.org/10.1016/j.cellimm.2017.11.010
PMid:29221689
8. Najar M, Bouhtit F, Melki R, Afif H, Hamal A, Fahmi H, Merimi M, Lagneaux L. Mesenchymal Stromal Cell-Based Therapy: New Perspectives and Challenges. J Clin Med. 2019;8:626. https://doi: 10.3390/jcm8050626
https://doi.org/10.3390/jcm8050626
PMid:31071990 PMCid:PMC6572531
9. Galipeau J,Sensébé L. Mesenchymal stromal cells: clinical challenges and therapeutic opportunities. Cell Stem Cell. 2018; 22 (6):824-833. https://doi:0.1016/j.stem.2018.05.004.
https://doi.org/10.1016/j.stem.2018.05.004
PMid:29859173 PMCid:PMC6434696
10. NajiA,EitokuM,FavierB,DeschaseauxF et al. Biological functions of mesenchymal stem cells and clinical implications. Cell Mol Life Sci. 2019;76 (17):3323-3348. https://doi: 10.1007/s00018-019-03125-1.
https://doi.org/10.1007/s00018-019-03125-1
PMid:31055643
11. Lotty A, Salama M, Zahran F, et. al. Characterization of Measenchymal Stem Cells Derived from Rat Bone Marrow and Adipose Tissue: a Comparative Study. Int J Stem Cells. 2014;7 (2):135-142.https://doi: 10.15283/ijsc.2014.7.2.135.
https://doi.org/10.15283/ijsc.2014.7.2.135
PMid:25473451 PMCid:PMC4249896
12. Viswanathan S, Shi Y, Galipeau J, Krampera M, Leblanc K, Martin I, et al. Mesenchymal stem versus stromal cells: International Society for Cell & Gene Therapy (ISCT (R)) Mesenchymal Stromal Cell committee position statement on nomenclature. Cytotherapy. 2019; 21:1019-24.https://doi: 10.1016/j.jcyt.2019.08.002.
https://doi.org/10.1016/j.jcyt.2019.08.002
PMid:31526643
13. Wilson A, Webster A, Genever P. Nomenclature and heterogeneity: consequences for the use of mesenchymal stem cells in regenerative medicine. Regen Med. 2019;14:595-611.https://doi: 10.2217/rme-2018-0145. Epub 2019 May 22
https://doi.org/10.2217/rme-2018-0145
PMid:31115266 PMCid:PMC7132560
14. Moll G, Hoogduijn MJ, Ankrum JA. Editorial: Safety, efficacy and mechanisms of action of mesenchymal stem cell therapies. Front Immunol. 2020;11:243. https://doi: 10.3389/fimmu.2020.00243.
https://doi.org/10.3389/fimmu.2020.00243
PMid:32133010 PMCid:PMC7040069
15. Peter RU, Panizzon RG, Seegenschmiedt MH. Diagnosis and treatment of cutaneous radiation injuries. In: Panizzon RG, Seegenschmiedt MH, editors. Radiation treatment and radiation reactions in dermatology. Berlin: Springer-Verlag Berlin Heidelberg.2015. p. 185-188. https://doi: 10.18632/aging.103932.
https://doi.org/10.18632/aging.103932
16. Lukomska B, Stanaszek L, Zuba-Surma E, Legosz P, Sarzynska S, Drela K. Challenges and controversies in human mesenchymal stem cell therapy. Stem Cells Int. 2019:9628536. https:// doi: 10.1155/2019/9628536. eCollection 2019.
https://doi.org/10.1155/2019/9628536
PMid:31093291 PMCid:PMC6481040
17. Shende P., Gupta H., Gaud R.S. Cytotherapy using stromal cells: Current and advance multi-treatment approaches. Biomed. Pharmacother. 2018;97:38-44. https:// doi: 10.1016/j.biopha.2017.10.127.
https://doi.org/10.1016/j.biopha.2017.10.127
PMid:29080456
18. Dominici M., Blanc K Le, Mueller I. et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 2006; 8 (4); 315-317. https://doi: 10.1080/14653240600855905.
https://doi.org/10.1080/14653240600855905
PMid:16923606
19. Wei J, Meng L, Hou X et al. Radiation-induced skin reactions: mechanism and treatment. Cancer Manag Res. 2019; 11: 167-177. https://doi: 10.2147/CMAR.S188655.
https://doi.org/10.2147/CMAR.S188655
PMid:30613164 PMCid:PMC6306060
20. Kumar S, Kolozsvary A, Kohl R, Lu M, Brown S, Kim JH. Radiation-induced skin injury in the animal model of scleroderma: implications for post-radiotherapy fibrosis. Radiat Oncol 2008; 3:40 https:// doi:10. 1186/1748-717X-3-40.
https://doi.org/10.1186/1748-717X-3-40
PMid:19025617 PMCid:PMC2599892
21. Pittenger MF. Mesenchymal stem cell perspective: cell biology to clinical progress. NPJRegen. Med. 2019;4:22-36. https:// doi: 10.1038/s41536-019-0083-6. eCollection 2019.
https://doi.org/10.1038/s41536-019-0083-6
PMid:31815001 PMCid:PMC6889290
22. Weiss ARR, Dahlke MH. Immunomodulation by mesenchymal stem cells (MSCs): mechanisms of action of living, apoptotic, and dead MSCs. Front. Immunol. 2019;10:1191.https:// doi: 10.3389/fimmu.2019.01191. eCollection 2019.
https://doi.org/10.3389/fimmu.2019.01191
PMid:31214172 PMCid:PMC6557979
23. Zhao L, Chen S, Yang P, Cao H, Li L. The role of mesenchymal stem cells in hematopoietic stem cell transplantation:prevention and treatment of graft-versus-host disease. Stem Cell Res Ther. 2019;10 (1):182 DOI: https:// doi:10.1186/s13287-019-1287-9
https://doi.org/10.1186/s13287-019-1287-9
PMid:31227011 PMCid:PMC6588914
24. Ullah M, Liu DD, Thakor AS. Mesenchymal Stromal Cell Homing: Mechanisms and Strategies for Improvement. iScience. 2019;15:421-438. https:// doi:10.1016/j.isci.2019.05.004. Epub 2019 May 9.
https://doi.org/10.1016/j.isci.2019.05.004
PMid:31121468 PMCid:PMC6529790
25. Hu KX, Sun QY, Gu M, Ai HS. The radiation protection and therapy effects of mesenchymal stem cells in mice with acute radiation injury. Br J Radiol. 2010;83 (985):52-58. https:// doi:10.1259/bjr/61042310
https://doi.org/10.1259/bjr/61042310
PMid:20139249 PMCid:PMC3487250
26. Lange C, Brunswig-Spickenheier B, Cappallo-Obermann H et al. Radiation rescue: mesenchymal stromal cells protect from lethal irradiation. PLoS One. 2011; 6:e14486. https:// doi:10.1371/journal.pone.0014486
https://doi.org/10.1371/journal.pone.0014486
PMid:21245929 PMCid:PMC3016319
27. Guerrouahen BS, Sidahmed H, Al Sulaiti A, Al Khulaifi M, Cugno C. Enhancing Mesenchymal Stromal Cell Immunomodulation for Treating Conditions Influenced by the Immune System. Stem Cells Int. 2020; 11:345-361. https://doi.org/10.1186/s13287-020-01855-9
https://doi.org/10.1186/s13287-020-01855-9
PMid:32771052 PMCid:PMC7414268
28. Zhou Y, Yamamoto Y, Xiao Z, Ochiya T. The Immunomodulatory Functions of Mesenchymal Stromal/Stem Cells Mediated via Paracrine Activity. J. Clin. Med. 2019;8:1025. https://doi: 10.3390/jcm8071025.
https://doi.org/10.3390/jcm8071025
PMid:31336889 PMCid:PMC6678920
29. Donnelly EH, Nemhauser JB, Smith JM, Kazzi ZN, Farfan EB, Chang AS, et al. Acute radiation syndrome: assessment and management. South Med J. 2010; 103:541-546. https://doi: 10.1097/SMJ.0b013e3181ddd571.
https://doi.org/10.1097/SMJ.0b013e3181ddd571
PMid:20710137
30. Hopewell JW. The Skin: Its Structure and Response to Ionizing Radiation. International Journal of Radiation Biology. 1990; 57:751-773. https:// doi: 10.1080/09553009014550911.
https://doi.org/10.1080/09553009014550911
PMid:1969905
31. Jimenez-Puerta GJ, Marchal JA, López-Ruiz E et al. Role of Mesenchymal Stromal Cells as Therapeutic Agents: Potential Mechanisms of Action and Implications in Their Clinical Use. J. Clin. Med. 2020; 9:445-461. https://doi:10.3390/jcm9020445
https://doi.org/10.3390/jcm9020445
PMid:32041213 PMCid:PMC7074225
32. Rittie L. Cellular mechanisms of skin repair in humans and other mammals. 2016;10 (2):103-120. https:// doi: 10.1007/s12079-016-0330-1.
https://doi.org/10.1007/s12079-016-0330-1
PMid:27170326 PMCid:PMC4882309
33. François S, Bensidhoum M, Mouiseddine M, et al. Local irradiation not only induces homing of human mesenchymal stem cells at exposed sites but promote their widespread engraftment to multiple to organs: A study of their quantitative distribution after irradiation damage. Stem Cells. 2006;24 (4):1020-1029 https:// doi: 10.1634/stemcells.2005-0260. Epub 2005 Dec 8.
https://doi.org/10.1634/stemcells.2005-0260
PMid:16339642
34. Hocking AM. The role of chemokines in mesenchymal stem cell homing to wounds. Adv Wound Care. 2015;4:623-630.https:// doi: 10.1089/wound.2014.0579
https://doi.org/10.1089/wound.2014.0579
PMid:26543676 PMCid:PMC4620518
35. Im G J, Kim J H, Wu HG, Hwang S J/ Effect of Mesenchymal Stem Cells and Platelet-Derived Growth Factor on the Healing of Radiation Induced Ulcer in Rats. Tissue Eng. Regen. Med. 2016;13 (1):78-90. http://dx.doi.org/10.1007/s13770-015-0055-x
https://doi.org/10.1007/s13770-015-0055-x
PMid:30603388 PMCid:PMC6170994
36. Isakson M, Blacam C, Whelan D, McArdle A, Clover AJP. Mesenchymal Stem Cells and Cutaneous Wound Healing: Current Evidence and Future Potential. Stem Cells International. 2015; 2015:831095. https:// doi:10.1155/2015/831095.
https://doi.org/10.1155/2015/831095
PMid:26106431 PMCid:PMC4461792

Uzlenkova N, Skorobogatova N, Kryvko A, Krasnoselsky M. The efficacy of cryopreserved ex vivo expanded rat bone marrow-derived multipotent mesenchymal stromal cells in the repair of radiation injuries in rats. Cell Organ Transpl. 2022; 10(1):10-16. Available from: https://doi.org/10.22494/cot.v10i1.139

 


Is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License (CC BY-NC 4.0)