Browse > Article
http://dx.doi.org/10.4062/biomolther.2017.260

Roles of Mesenchymal Stem Cells in Tissue Regeneration and Immunomodulation  

Ayala-Cuellar, Ana Patricia (Laboratory of Biochemistry and Immunology, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University)
Kang, Ji-Houn (Laboratory of Internal Medicine, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University)
Jeung, Eui-Bae (Laboratory of Biochemistry and Molecular Biology, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University)
Choi, Kyung-Chul (Laboratory of Biochemistry and Immunology, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University)
Publication Information
Biomolecules & Therapeutics / v.27, no.1, 2019 , pp. 25-33 More about this Journal
Abstract
Mesenchymal stem cells are classified as multipotent stem cells, due to their capability to transdifferentiate into various lineages that develop from mesoderm. Their popular appeal as cell-based therapy was initially based on the idea of their ability to restore tissue because of their differentiation potential in vitro; however, the lack of evidence of their differentiation to target cells in vivo led researchers to focus on their secreted trophic factors and their role as potential powerhouses on regulation of factors under different immunological environments and recover homeostasis. To date there are more than 800 clinical trials on humans related to MSCs as therapy, not to mention that in animals is actively being applied as therapeutic resource, though it has not been officially approved as one. But just as how results from clinical trials are important, so is to reveal the biological mechanisms involved on how these cells exert their healing properties to further enhance the application of MSCs on potential patients. In this review, we describe characteristics of MSCs, evaluate their benefits as tissue regenerative therapy and combination therapy, as well as their immunological properties, activation of MSCs that dictate their secreted factors, interactions with other immune cells, such as T cells and possible mechanisms and pathways involved in these interactions.
Keywords
Mesenchymal stem cells; Immunomodulation; Regenerative medicine; Toll-like receptor; Prostaglandin E2; T regulators;
Citations & Related Records
연도 인용수 순위
  • Reference
1 Bartholomew, A., Sturgeon, C., Siatskas, M., Ferrer, K., McIntosh, K., Patil, S., Hardy, W., Devine, S., Ucker, D., Deans, R., Moseley, A. and Hoffman, R. (2002) Mesenchymal stem cells suppress lymphocyte proliferation in vitro and prolong skin graft survival in vivo. Exp. Hematol. 30, 42-48.   DOI
2 Bonab, M. M., Alimoghaddam, K., Talebian, F., Ghaffari, S. H., Ghavamzadeh, A. and Nikbin, B. (2006) Aging of mesenchymal stem cell in vitro. BMC Cell Biol. 7, 14.   DOI
3 Boomsma, R. A. and Geenen, D. L. (2012) Mesenchymal stem cells secrete multiple cytokines that promote angiogenesis and have contrasting effects on chemotaxis and apoptosis. PLoS ONE 7, e35685.   DOI
4 Lei, J., Wang, Z., Hui, D., Yu, W., Zhou, D., Xia, W., Chen, C., Zhang, Q., Wang, Z., Zhang, Q. and Xiang, A. P. (2011) Ligation of TLR2 and TLR4 on murine bone marrow-derived mesenchymal stem cells triggers differential effects on their immunosuppressive activity. Cell. Immunol. 271, 147-156.   DOI
5 Lim, J. Y., Im, K. I., Lee, E. S., Kim, N., Nam, Y. S., Jeon, Y. W. and Cho, S. G. (2016) Enhanced immunoregulation of mesenchymal stem cells by IL-10-producing type 1 regulatory T cells in collagen-induced arthritis. Sci. Rep. 6, 26851.   DOI
6 Liu, Y. L., Liu, W. H., Sun, J., Hou, T. J., Liu, Y. M., Liu, H. R., Luo, Y. H., Zhao, N. N., Tang, Y. and Deng, F. M. (2014) Mesenchymal stem cell-mediated suppression of hypertrophic scarring is p53 dependent in a rabbit ear model. Stem Cell Res. Ther. 5, 136.   DOI
7 Lo, B. and Parham, L. (2009) Ethical issues in stem cell research. Endocr. Rev. 30, 204-213.   DOI
8 Ma, S., Chen, X., Wang, L., Wei, Y., Ni, Y., Chu, Y., Liu, Y., Zhu, H., Zheng, R. and Zhang, Y. (2017) Repairing effects of ICAM-1-expressing mesenchymal stem cells in mice with autoimmune thyroiditis. Exp. Ther. Med. 13, 1295-1302.   DOI
9 Mahla, R. S. (2016) Stem cells applications in regenerative medicine and disease therapeutics. Int. J. Cell Biol. 2016, 6940283.   DOI
10 Matsui, F., Babitz, S. A., Rhee, A., Hile, K. L., Zhang, H. and Meldrum, K. K. (2017) Mesenchymal stem cells protect against obstruction-induced renal fibrosis by decreasing STAT3 activation and STAT3-dependent MMP-9 production. Am. J. Physiol. Renal Physiol. 312, F25-F32.   DOI
11 Matsui, F. and Meldrum, K. K. (2012) The role of the Janus kinase family/signal transducer and activator of transcription signaling pathway in fibrotic renal disease. J. Surg. Res. 178, 339-345.   DOI
12 Xiong, H., Bai, C., Wu, S., Gao, Y., Lu, T., Hu, Q., Guan, W. and Ma, Y. (2014) Biological characterization of mesenchymal stem cells from bovine umbilical cord. Animal Cells and Systems 18, 59-67.   DOI
13 Ulrich, D., Muralitharan, R. and Gargett, C. E. (2013) Toward the use of endometrial and menstrual blood mesenchymal stem cells for cell-based therapies. Expert Opin. Biol. Ther. 13, 1387-1400.   DOI
14 Ullah, I., Subbarao, R. B. and Rho, G. J. (2015) Human mesenchymal stem cells - current trends and future prospective. Biosci. Rep. 35, e00191.   DOI
15 Vidal, M. A., Robinson, S. O., Lopez, M. J., Paulsen, D. B., Borkhsenious, O., Johnson, J. R., Moore, R. M. and Gimble, J. M. (2008) Comparison of chondrogenic potential in equine mesenchymal stromal cells derived from adipose tissue and bone marrow. Vet. Surg. 37, 713-724.   DOI
16 Volk, S. W. and Theoret, C. (2013) Translating stem cell therapies: the role of companion animals in regenerative medicine. Wound Repair Regen. 21, 382-394.   DOI
17 Wipff, P. J. and Hinz, B. (2008) Integrins and the activation of latent transforming growth factor beta1 - an intimate relationship. Eur. J. Cell Biol. 87, 601-615.   DOI
18 Zhu, W. and Liang, M. (2015) Periodontal ligament stem cells: current status, concerns, and future prospects. Stem Cells Int. 2015, 972313.   DOI
19 Burr, S. P., Dazzi, F. and Garden, O. A. (2013) Mesenchymal stromal cells and regulatory T cells: the Yin and Yang of peripheral tolerance? Immunol. Cell Biol. 91, 12-18.   DOI
20 Brown, S. G., Harman, R. J. and Black, L. L. (2012) Adipose-derived stem cell therapy for severe muscle tears in working German shepherds: two case reports. Stem Cell Discovery 2, 41-44.   DOI
21 Byeon, J. S., Lee, J., Kim, D. H., Lee, G.-B., Kim, H.-R., Gu, N.-Y., Cho, I.-S. and Cha, S.-H. (2016) Canine mesenchymal stem cells immunomodulate atopic dermatitis through the induction of regulatory T cells in an ex vivo experimental study. J. Prev. Vet. Med. 40, 12-21.   DOI
22 Caplan, A. I. (1991) Mesenchymal stem cells. J. Orthop. Res. 9, 641-650.   DOI
23 Carrade Holt, D. D., Wood, J. A., Granick, J. L., Walker, N. J., Clark, K. C. and Borjesson, D. L. (2014) Equine mesenchymal stem cells inhibit T cell proliferation through different mechanisms depending on tissue source. Stem Cells Dev. 23, 1258-1265.   DOI
24 de Mattos Carvalho, A., Alves, A. L. G., de Oliveira, P. G. G., Cisneros Alvarez, L. E., Amorim, R. L., Hussni, C. A. and Deffune, E. (2011) Use of adipose tissue-derived mesenchymal stem cells for experimental tendinitis therapy in equines. J. Equine Vet. Sci. 31, 26-34.   DOI
25 Case, J. B., Palmer, R., Valdes-Martinez, A., Egger, E. L. and Haussler, K. K. (2013) Gastrocnemius tendon strain in a dog treated with autologous mesenchymal stem cells and a custom orthosis. Vet. Surg. 42, 355-360.   DOI
26 Chen, L. B., Jiang, X. B. and Yang, L. (2004) Differentiation of rat marrow mesenchymal stem cells into pancreatic islet beta-cells. World J. Gastroenterol. 10, 3016-3020.   DOI
27 Cyranoski, D. (2013) Stem cells boom in vet clinics. Nature 496, 148-149.   DOI
28 Nordgren, T. M., Bailey, K. L., Heires, A. J., Katafiasz, D. and Romberger, D. J. (2018) Effects of agricultural organic dusts on human lung-resident mesenchymal stem (stromal) cell function. Toxicol. Sci. 162, 635-644.   DOI
29 Mrozik, K. M., Zilm, P. S., Bagley, C. J., Hack, S., Hoffmann, P., Gronthos, S. and Bartold, P. M. (2010) Proteomic characterization of mesenchymal stem cell-like populations derived from ovine periodontal ligament, dental pulp, and bone marrow: analysis of differentially expressed proteins. Stem Cells De. 19, 1485-1499.   DOI
30 Najimi, M., Berardis, S., El-Kehdy, H., Rosseels, V., Evraerts, J., Lombard, C., El Taghdouini, A., Henriet, P., van Grunsven, L. and Sokal, E. M. (2017) Human liver mesenchymal stem/progenitor cells inhibit hepatic stellate cell activation: in vitro and in vivo evaluation. Stem Cell Res. Ther. 8, 131.   DOI
31 Otabe, K., Muneta, T., Kawashima, N., Suda, H., Tsuji, K. and Sekiya, I. (2012) Comparison of gingiva, dental pulp, and periodontal ligament cells from the standpoint of mesenchymal stem cell properties. Cell Med. 4, 13-21.   DOI
32 Plumas, J., Chaperot, L., Richard, M. J., Molens, J. P., Bensa, J. C. and Favrot, M. C. (2005) Mesenchymal stem cells induce apoptosis of activated T cells. Leukemia 19, 1597-1604.   DOI
33 Raabe, O., Shell, K., Goessl, A., Crispens, C., Delhasse, Y., Eva, A., Scheiner-Bobis, G., Wenisch, S. and Arnhold, S. (2013) Effect of extracorporeal shock wave on proliferation and differentiation of equine adipose tissue-derived mesenchymal stem cells in vitro. Am. J. Stem Cells. 8, 2, 62-73.
34 Rawlings, J. S., Rosler, K. M. and Harrison, D. A. (2004) The JAK/STAT signaling pathway. J Cell Sci. 117, 1281-1283.   DOI
35 Fallarino, F., Grohmann, U., Vacca, C., Bianchi, R., Orabona, C., Spreca, A., Fioretti, M. C. and Puccetti, P. (2002) T cell apoptosis by tryptophan catabolism. Cell Death Differ. 9, 1069-1077.   DOI
36 Domergue, S., Bony, C., Maumus, M., Toupet, K., Frouin, E., Rigau, V., Vozenin, M. C., Magalon, G., Jorgensen, C. and Noel, D. (2016) Comparison between stromal vascular fraction and adipose mesenchymal stem cells in remodeling hypertrophic scars. PLoS ONE 11, e0156161.   DOI
37 Dominici, M., Le Blanc, K., Mueller, I., Slaper-Cortenbach, I., Marini, F., Krause, D., Deans, R., Keating, A., Prockop, D. and Horwitz, E. (2006) Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 8, 315-317.   DOI
38 Engela, A. U., Baan, C. C., Dor, F. J., Weimar, W. and Hoogduijn, M. J. (2012) On the interactions between mesenchymal stem cells and regulatory T cells for immunomodulation in transplantation. Front. Immunol. 3, 126.   DOI
39 Engela, A. U., Baan, C. C., Peeters, A. M., Weimar, W. and Hoogduijn, M. J. (2013) Interaction between adipose tissue-derived mesenchymal stem cells and regulatory T-cells. Cell Transplant. 22, 41-54.   DOI
40 English, K. and Mahon, B. P. (2011) Allogeneic mesenchymal stem cells: agents of immune modulation. J. Cell. Biochem. 112, 1963-1968.   DOI
41 Fei, X., Jiang, S., Zhang, S., Li, Y., Ge, J., He, B., Goldstein, S. and Ruiz, G. (2013) Isolation, culture, and identification of amniotic fluid-derived mesenchymal stem cells. Cell Biochem. Biophys. 67, 689-694.   DOI
42 Friedenstein, A. J., Chailakhjan, R. K. and Lalykina, K. S. (1970) The development of fibroblast colonies in monolayer cultures of guineapig bone marrow and spleen cells. Cell Tissue Kinet. 3, 393-403.
43 Kim, C. H., Lee, J. H., Won, J. H. and Cho, M. K. (2011) Mesenchymal stem cells improve wound healing in vivo via early activation of matrix metalloproteinase-9 and vascular endothelial growth factor. J. Korean Med. Sci. 26, 726-733.   DOI
44 Kaewsuwan, S., Song, S. Y., Kim, J. H. and Sung, J. H. (2012) Mimicking the functional niche of adipose-derived stem cells for regenerative medicine. Expert Opin. Biol. Ther. 12, 1575-1588.   DOI
45 Kalinski, P. (2012) Regulation of immune responses by prostaglandin E2. J. Immunol. 188, 21-28.   DOI
46 Kang, J. W., Kang, K. S., Koo, H. C., Park, J. R., Choi, E. W. and Park, Y. H. (2008) Soluble factors-mediated immunomodulatory effects of canine adipose tissue-derived mesenchymal stem cells. Stem Cells Dev. 17, 681-693.   DOI
47 Koch, T. G., Heerkens, T., Thomsen, P. D. and Betts, D. H. (2007) Isolation of mesenchymal stem cells from equine umbilical cord blood. BMC Biotechnol. 7, 26.   DOI
48 Kim, H. S., Kim, K. H., Kim, S. H., Kim, Y. S., Koo, K. T., Kim, T. I., Seol, Y. J., Ku, Y., Rhyu, I. C., Chung, C. P. and Lee, Y. M. (2010) Immunomodulatory effect of canine periodontal ligament stem cells on allogenic and xenogenic peripheral blood mononuclear cells. J. Periodontal. Implant. Sci. 40, 265-270.   DOI
49 Kim, H. W., Song, W. J., Li, Q., Han, S. M., Jeon, K. O., Park, S. C., Ryu, M. O., Chae, H. K., Kyeong, K. and Youn, H. Y. (2016) Canine adipose tissue-derived mesenchymal stem cells ameliorate severe acute pancreatitis by regulating T cells in rats. J. Vet. Sci. 17, 539-548.   DOI
50 Kisiel, A. H., McDuffee, L. A., Masaoud, E., Bailey, T. R., Esparza Gonzalez, B. P. and Nino-Fong, R. (2012) 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.   DOI
51 Ryan, J. M., Barry, F., Murphy, J. M. and Mahon, B. P. (2007) Interferon-gamma does not break, but promotes the immunosuppressive capacity of adult human mesenchymal stem cells. Clin. Exp. Immunol. 149, 353-363.   DOI
52 Reiter, J., Drummond, S., Sammour, I., Huang, J., Florea, V., Dornas, P., Hare, J. M., Rodrigues, C. O. and Young, K. C. (2017) Stromal derived factor-1 mediates the lung regenerative effects of mesenchymal stem cells in a rodent model of bronchopulmonary dysplasia. Respir. Res. 18, 137.   DOI
53 Ren, H., Sang, Y., Zhang, F., Liu, Z., Qi, N. and Chen, Y. (2016) Comparative analysis of human mesenchymal stem cells from umbilical cord, dental pulp, and menstrual blood as sources for cell therapy. Stem Cells Int. 2016, 3516574.
54 Rhee, K. J., Lee, J. I. and Eom, Y. W. (2015) Mesenchymal Stem Cell-Mediated Effects of Tumor Support or Suppression. Int. J. Mol. Sci. 16, 30015-30033.   DOI
55 Sato, K., Ozaki, K., Oh, I., Meguro, A., Hatanaka, K., Nagai, T., Muroi, K. and Ozawa, K. (2007) Nitric oxide plays a critical role in suppression of T-cell proliferation by mesenchymal stem cells. Blood 109, 228-234.   DOI
56 Schlosser, S., Dennler, C., Schweizer, R., Eberli, D., Stein, J. V., Enzmann, V., Giovanoli, P., Erni, D. and Plock, J. A. (2012) Paracrine effects of mesenchymal stem cells enhance vascular regeneration in ischemic murine skin. Microvasc. Res. 83, 267-275.   DOI
57 Gibson, M., Brown, S. G. and Brown, N. O. (2017) Semitendinosus myopathy and treatment with adipose-derived stem cells in working German shepherd police dogs. Can. Vet. J. 58, 241-246.
58 Schuh, E. M., Friedman, M. S., Carrade, D. D., Li, J., Heeke, D., Oyserman, S. M., Galuppo, L. D., Lara, D. J., Walker, N. J., Ferraro, G. L., Owens, S. D. and Borjesson, D. L. (2009) Identification of variables that optimize isolation and culture of multipotent mesenchymal stem cells from equine umbilical-cord blood. Am. J. Vet. Res. 70, 1526-1535.   DOI
59 Gaafar, T., Attia, W., Mahmoud, S., Sabry, D., Aziz, O. A., Rasheed, D. and Hamza, H. (2017) Cardioprotective effects of wharton jelly derived mesenchymal stem cell transplantation in a rodent model of myocardial injury. Int. J. Stem Cells 10, 48-59.   DOI
60 Ganguly, P., El-Jawhari, J. J., Giannoudis, P. V., Burska, A. N., Ponchel, F. and Jones, E. A. (2017) Age-related changes in bone marrow mesenchymal stromal cells: a potential impact on osteoporosis and osteoarthritis development. Cell Transplant. 26, 1520-1529.   DOI
61 Glennie, S., Soeiro, I., Dyson, P. J., Lam, E. W. and Dazzi, F. (2005) Bone marrow mesenchymal stem cells induce division arrest anergy of activated T cells. Blood 105, 2821-2827.   DOI
62 Guercio, A., Di Marco, P., Casella, S., Cannella, V., Russotto, L., Purpari, G., Di Bella, S. and Piccione, G. (2012) Production of canine mesenchymal stem cells from adipose tissue and their application in dogs with chronic osteoarthritis of the humeroradial joints. Cell Biol. Int. 36, 189-194.   DOI
63 Hakki, S. S., Turac, G., Bozkurt, S. B., Kayis, S. A., Hakki, E. E., Sahin, E., Subasi, C. and Karaoz, E. (2017) Comparison of different sources of mesenchymal stem cells: palatal versus lipoaspirated adipose tissue. Cells Tissues Organs 204, 228-240.   DOI
64 Han, S. M., Kim, H. T., Kim, K. W., Jeon, K. O., Seo, K. W., Choi, E. W. and Youn, H. Y. (2015) CTLA4 overexpressing adipose tissuederived mesenchymal stem cell therapy in a dog with steroid-refractory pemphigus foliaceus. BMC Vet. Res. 11, 49.   DOI
65 Jacobs, S. A., Roobrouck, V. D., Verfaillie, C. M. and Van Gool, S. W. (2013) Immunological characteristics of human mesenchymal stem cells and multipotent adult progenitor cells. Immunol. Cell Biol. 91, 32-39.   DOI
66 Le Blanc, K., Tammik, L., Sundberg, B., Haynesworth, S. E. and Ringden, O. (2003) Mesenchymal stem cells inhibit and stimulate mixed lymphocyte cultures and mitogenic responses independently of the major histocompatibility complex. Scand. J. Immunol. 57, 11-20.   DOI
67 Kondo, M., Yamaoka, K., Sakata, K., Sonomoto, K., Lin, L., Nakano, K. and Tanaka, Y. (2015) Contribution of the Interleukin-6/STAT-3 signaling pathway to chondrogenic differentiation of human mesenchymal stem cells. Arthritis Rheumatol. 67, 1250-1260.   DOI
68 Kopen, G. C., Prockop, D. J. and Phinney, D. G. (1999) Marrow stromal cells migrate throughout forebrain and cerebellum, and they differentiate into astrocytes after injection into neonatal mouse brains. Proc. Natl. Acad. Sci. U.S.A. 96, 10711-10716.   DOI
69 Krampera, M., Galipeau, J., Shi, Y., Tarte, K. and Sensebe, L. (2013) Immunological characterization of multipotent mesenchymal stromal cells--The International Society for Cellular Therapy (ISCT) working proposal. Cytotherapy 15, 1054-1061.   DOI
70 Lawson, J., Elliott, J., Wheeler-Jones, C., Syme, H. and Jepson, R. (2015) Renal fibrosis in feline chronic kidney disease: known mediators and mechanisms of injury. Vet. J. 203, 18-26.   DOI
71 Lee, J. Y., Uzuka, Y., Tanabe, S. and Sarashina, T. (2004) Prevalence of thyroglobulin autoantibodies detected by enzyme-linked immunosorbent assay of canine serum in hypothyroid, obese and healthy dogs in Japan. Res. Vet. Sci. 76, 129-132.   DOI
72 Lee, W. S., Suzuki, Y., Graves, S. S., Iwata, M., Venkataraman, G. M., Mielcarek, M., Peterson, L. J., Ikehara, S., Torok-Storb, B. and Storb, R. (2011) Canine bone marrow-derived mesenchymal stromal cells suppress alloreactive lymphocyte proliferation in vitro but fail to enhance engraftment in canine bone marrow transplantation. Biol. Blood Marrow Transplant. 17, 465-475.   DOI
73 Sousa, M. G., Paulino-Junior, D., Pascon, J. P., Pereira-Neto, G. B., Carareto, R., Champion, T. and Camacho, A. A. (2011) Cardiac function in dogs with chronic Chagas cardiomyopathy undergoing autologous stem cell transplantation into the coronary arteries. Can. Vet. J. 52, 869-874.
74 Shabbir, A., Zisa, D., Lin, H., Mastri, M., Roloff, G., Suzuki, G. and Lee, T. (2010) Activation of host tissue trophic factors through JAK-STAT3 signaling: a mechanism of mesenchymal stem cell-mediated cardiac repair. Am. J. Physiol. Heart Circ. Physiol. 299, H1428-H1438.   DOI
75 Sharma, R. R., Pollock, K., Hubel, A. and McKenna, D. (2014) Mesenchymal stem or stromal cells: a review of clinical applications and manufacturing practices. Transfusion 54, 1418-1437.   DOI
76 Singer, N. G. and Caplan, A. I. (2011) Mesenchymal stem cells: mechanisms of inflammation. Annu. Rev. Pathol. 6, 457-478.   DOI
77 Squillaro, T., Peluso, G. and Galderisi, U. (2016) Clinical trials with mesenchymal stem cells: an update. Cell Transplant. 25, 829-848.   DOI
78 Tidd, N., Michelsen, J., Hilbert, B. and Quinn, J. C. (2017) Minicircle mediated gene delivery to canine and equine mesenchymal stem cells. Int. J. Mol. Sci. 18, E819.   DOI
79 Strioga, M., Viswanathan, S., Darinskas, A., Slaby, O. and Michalek, J. (2012) Same or not the same? Comparison of adipose tissue-derived versus bone marrow-derived mesenchymal stem and stromal cells. Stem Cells Dev. 21, 2724-2752.   DOI
80 Teixeira, F. G., Panchalingam, K. M., Anjo, S. I., Manadas, B., Pereira, R., Sousa, N., Salgado, A. J. and Behie, L. A. (2015) Do hypoxia/normoxia culturing conditions change the neuroregulatory profile of Wharton Jelly mesenchymal stem cell secretome? Stem Cell Res. Ther. 6, 133.   DOI
81 Bahamondes, F., Flores, E., Cattaneo, G., Bruna, F. and Conget, P. (2017) Omental adipose tissue is a more suitable source of canine Mesenchymal stem cells. BMC Vet. Res. 13, 166.   DOI
82 Abouelkheir, M., Eltantawy, D. A., Saad, M.-A., Abdelrahman, K. M., Sobh, M.-A., Lotfy, A. and Sobh, M. A. (2016) Mesenchymal stem cells versus their conditioned medium in the treatment of cisplatin-induced acute kidney injury: evaluation of efficacy and cellular side effects. Int. J. Clin. Exp. Med. 9, 23222-23234.
83 Alcayaga-Miranda, F., Cuenca, J., Luz-Crawford, P., Aguila-Diaz, C., Fernandez, A., Figueroa, F. E. and Khoury, M. (2015) Characterization of menstrual stem cells: angiogenic effect, migration and hematopoietic stem cell support in comparison with bone marrow mesenchymal stem cells. Stem Cell Res. Ther. 6, 32.   DOI
84 Arutyunyan, I., Elchaninov, A., Makarov, A. and Fatkhudinov, T. (2016) Umbilical Cord as Prospective Source for Mesenchymal Stem Cell-Based Therapy. Stem Cells Int. 2016, 6901286.
85 Bai, C., Gao, Y., Li, Q., Feng, Y., Yu, Y., Meng, G., Zhang, M. and Guan, W. (2015) Differentiation of chicken umbilical cord mesenchymal stem cells into beta-like pancreatic islet cells. Artif. Cells Nanomed. Biotechnol. 43, 106-111.   DOI