Comparison of therapeutic effects of mesenchymal stem cells from umbilical cord and bone marrow in the treatment of type 1 diabetes

Wei Zhang, Qing Ling, Bin Wang, Kai Wang, Jianbo Pang, Jing Lu, Yan Bi, Dalong Zhu, Wei Zhang, Qing Ling, Bin Wang, Kai Wang, Jianbo Pang, Jing Lu, Yan Bi, Dalong Zhu

Abstract

Background: The therapeutic potential of mesenchymal stem cells (MSCs) in type 1 diabetes (T1D) has been demonstrated in both preclinical and clinical studies. MSCs that have been used in research on T1D are derived from various tissue sources, with bone marrow (BM) and umbilical cord (UC) tissues being the most commonly used. However, the influence of tissue origin on the functional properties and therapeutic effects of MSCs in T1D remains unclear. This study aimed to compare the therapeutic efficacy of UC-MSCs and BM-MSCs in a mouse model of T1D as well as in patients with T1D.

Methods: In non-obese diabetic (NOD) mice, the development of diabetes was accelerated by streptozotocin injections. Thereafter, diabetic mice were randomized and treated intravenously with UC-MSCs, BM-MSCs or phosphate-buffered saline as a control. Blood glucose and serum insulin were measured longitudinally after transplantation. At 14 days post-transplantation, pancreatic tissues were collected to assess insulitis and the β-cell mass. Flow cytometry was performed to evaluate the composition of T lymphocytes in the spleen and pancreatic lymph nodes of the NOD mice. In our retrospective study of patients with T1D, 28 recipients who received insulin therapy alone or a single transplantation of UC-MSCs or BM-MSCs were enrolled. The glycaemic control and β-cell function of the patients during the first year of follow-up were compared.

Results: In NOD mice, UC-MSC and BM-MSC transplantation showed similar effects on decreasing blood glucose levels and preserving β cells. The regulation of islet autoimmunity was examined, and no significant difference between UC-MSCs and BM-MSCs was observed in the attenuation of insulitis, the decrease in T helper 17 cells or the increase in regulatory T cells. In patients with T1D, MSC transplantation markedly lowered haemoglobin A1c (HbA1c) levels and reduced insulin doses compared to conventional insulin therapy. However, the therapeutic effects were comparable between UC-MSCs and BM-MSCs, and they also exerted similar effects on the endogenous β-cell function in the patients.

Conclusion: In conclusion, both UC-MSCs and BM-MSCs exhibited comparable therapeutic effects on improving glycaemic control and preserving β-cell function in T1D. Considering their abundance and higher cell yields, UC-MSCs appear to be more promising than BM-MSCs in clinical applications. Trial registration NCT02763423. Registered on May 5, 2016-Retrospectively registered, https://www.

Clinicaltrials: gov/ .

Keywords: Bone marrow-derived mesenchymal stem cells; Cell transplantation; Type 1 diabetes; Umbilical cord-derived mesenchymal stem cells.

Conflict of interest statement

The authors declare that they have no competing interests.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
Characterization of BM-MSCs and UC-MSCs. A Representative micrographs of BM-MSCs and UC-MSCs observed under light microscopy. Scale bar, 200 μm. B Expression of CD14, CD73, CD19, CD90, CD34, CD105, CD45 and HLA-DR in BM-MSCs and UC-MSCs analysed by flow cytometry. C Representative micrographs of adipogenesis identified by Oil Red O staining, and osteogenesis identified by Alizarin Red staining of BM-MSCs and UC-MSCs. Scale bar, 100 μm
Fig. 2
Fig. 2
Effects of BM-MSCs or UC-MSCs infusion on body weights and blood glucose levels in NOD mice. A The treatment schedule for STZ and MSC transplantation. Body weights (B) and blood glucose levels (C) were measured twice a week after MSC transplantation for 14 days (N = 13 in each group). Data are presented as mean ± SEM. *P < 0.05 versus the control group
Fig. 3
Fig. 3
Effects of BM-MSCs or UC-MSCs infusion on β-cell mass and insulin production. A Representative micrographs of insulin immunohistochemical staining showing β cells preserved in islets at 14 days post-transplantation. Scale bar, 100 μm. B The insulin-positive staining areas (%) in pancreatic islets were quantified using the Image-Pro Plus 6.0. Four slides per mouse (four mice per group) were analysed, and at least 20 islets from the pancreatic tissue of each mouse were evaluated. C Random serum insulin concentration (N = 4–5). Data are presented as mean ± SEM. *P < 0.05 versus the control group
Fig. 4
Fig. 4
Effects of BM-MSCs or UC-MSCs infusion on insulitis in NOD mice. A Representative micrographs of H&E staining of pancreas at 14 days post-transplantation. Scale bar, 100 μm. B The percentage of islets in each of the infiltration categories (score 0, no insulitis; score 1, leukocyte infiltration ≤ 25%; score 2, leukocyte infiltration > 25%, but ≤ 50%; score 3, leukocyte infiltration > 50%; score 4, leukocyte infiltration 100% and β-cell destruction). C Insulitis score in pancreatic islets. Four slides per mouse (four mice per group) were analysed, and at least 20 islets from the pancreatic tissue of each mouse were evaluated. Data are presented as mean ± SEM. *P < 0.05 versus the control group
Fig. 5
Fig. 5
Effects of BM-MSCs or UC-MSCs infusion on T cell proportion in NOD mice. Percentage of CD8 + T cells (A) and CD4 + T cells (B) in gated CD3 + T cell population in the spleen and PLNs from NOD mice killed at 14 days post-treatment. Representative cytofluorometric dot plots (C) and summary data (D) of Th17 in gated CD4 + T cell population in the spleen and PLNs from NOD mice killed at 14 days post-treatment. Representative cytofluorometric dot plots (E) and summary data (F) of Tregs in gated CD4 + T cell population in the spleen and PLNs from NOD mice killed at 14 days post-treatment (N = 4–6). PLN, pancreatic lymph node
Fig. 6
Fig. 6
Change in HbA1c, exogenous insulin dosage and C-peptide in patients with T1D at 1-year follow-up. Rate of change in HbA1c (A), doses of daily insulin (B), FCP (C) and PCP (D) of the control group and MSC-treated group between baseline and 1-year follow-up. Rate of change in HbA1c (E), doses of daily insulin (F), FCP (G) and PCP (H) of the BM-MSC subgroup and UC-MSC subgroup between baseline and 1-year follow-up. Data are presented as mean ± SEM. *P < 0.05 versus the control group. FCP, fasting C-peptide; PCP, postprandial C-peptide

References

    1. Perkins BA, Sherr JL, Mathieu C. Type 1 diabetes glycemic management: insulin therapy, glucose monitoring, and automation. Science (New York, NY) 2021;373(6554):522–527. doi: 10.1126/science.abg4502.
    1. Farkhad NK, Mahmoudi A, Mahdipour E. How similar are human mesenchymal stem cells derived from different origins? A review of comparative studies. Curr Stem Cell Res Ther. 2021;16(8):980–993. doi: 10.2174/1574888X16666210302151823.
    1. Song N, Scholtemeijer M, Shah K. Mesenchymal stem cell immunomodulation: mechanisms and therapeutic potential. Trends Pharmacol Sci. 2020;41(9):653–664. doi: 10.1016/j.tips.2020.06.009.
    1. Galipeau J, Sensebe L. Mesenchymal stromal cells: clinical challenges and therapeutic opportunities. Cell Stem Cell. 2018;22(6):824–833. doi: 10.1016/j.stem.2018.05.004.
    1. Krampera M, Le Blanc K. Mesenchymal stromal cells: putative microenvironmental modulators become cell therapy. Cell Stem Cell. 2021;28(10):1708–1725. doi: 10.1016/j.stem.2021.09.006.
    1. Jurewicz M, Yang S, Augello A, Godwin JG, Moore RF, Azzi J, et al. Congenic mesenchymal stem cell therapy reverses hyperglycemia in experimental type 1 diabetes. Diabetes. 2010;59(12):3139–3147. doi: 10.2337/db10-0542.
    1. Kota DJ, Wiggins LL, Yoon N, Lee RH. TSG-6 produced by hMSCs delays the onset of autoimmune diabetes by suppressing Th1 development and enhancing tolerogenicity. Diabetes. 2013;62(6):2048–2058. doi: 10.2337/db12-0931.
    1. Khatri R, Petry SF, Linn T. Intrapancreatic MSC transplantation facilitates pancreatic islet regeneration. Stem Cell Res Ther. 2021;12(1):121. doi: 10.1186/s13287-021-02173-4.
    1. Li L, Hui H, Jia X, Zhang J, Liu Y, Xu Q, et al. Infusion with human bone marrow-derived mesenchymal stem cells improves beta-cell function in patients and non-obese mice with severe diabetes. Sci Rep. 2016;6:37894. doi: 10.1038/srep37894.
    1. Refaie AF, Elbassiouny BL, Kloc M, Sabek OM, Khater SM, Ismail AM, et al. From mesenchymal stromal/stem cells to insulin-producing cells: immunological considerations. Front Immunol. 2021;12:690623. doi: 10.3389/fimmu.2021.690623.
    1. Gilkeson GA-O. Safety and efficacy of mesenchymal stromal cells and other cellular therapeutics in rheumatic diseases in 2022: a review of what we know so far. Arthritis Rheumatol. 2022 doi: 10.1002/art.42081.
    1. Piekarska K, Urban-Wojciuk Z, Kurkowiak M, Pelikant-Malecka I, Schumacher A, Sakowska J, et al. Mesenchymal stem cells transfer mitochondria to allogeneic Tregs in an HLA-dependent manner improving their immunosuppressive activity. Nat Commun. 2022;13(1):856. doi: 10.1038/s41467-022-28338-0.
    1. Forbes S, Bond AR, Thirlwell KL, Burgoyne P, Samuel K, Noble J, et al. Human umbilical cord perivascular cells improve human pancreatic islet transplant function by increasing vascularization. Sci Transl Med. 2020;12(526):eaan5907. doi: 10.1126/scitranslmed.aan5907.
    1. Sun YL, Shang LR, Liu RH, Li XY, Zhang SH, Ren YK, et al. Therapeutic effects of menstrual blood-derived endometrial stem cells on mouse models of streptozotocin-induced type 1 diabetes. World J Stem Cells. 2022;14(1):104–116. doi: 10.4252/wjsc.v14.i1.104.
    1. Wang L, Liu T, Liang R, Wang G, Liu Y, Zou J, et al. Mesenchymal stem cells ameliorate beta cell dysfunction of human type 2 diabetic islets by reversing beta cell dedifferentiation. EBioMedicine. 2020;51:102615. doi: 10.1016/j.ebiom.2019.102615.
    1. Tang S, Zhang M, Zeng S, Huang Y, Qin M, Nasri U, et al. Reversal of autoimmunity by mixed chimerism enables reactivation of beta cells and transdifferentiation of alpha cells in diabetic NOD mice. Proc Natl Acad Sci U S A. 2020;117(49):31219–31230. doi: 10.1073/pnas.2012389117.
    1. Cho J, D'Antuono M, Glicksman M, Wang J, Jonklaas J. A review of clinical trials: mesenchymal stem cell transplant therapy in type 1 and type 2 diabetes mellitus. Am J Stem Cells. 2018;7(4):82–93.
    1. Pixley JS. Mesenchymal stem cells to treat type 1 diabetes. Biochim Biophys Acta Mol Basis Dis. 2020;1866(4):165315. doi: 10.1016/j.bbadis.2018.10.033.
    1. Li Y, Wang F, Liang H, Tang D, Huang M, Zhao J, et al. Efficacy of mesenchymal stem cell transplantation therapy for type 1 and type 2 diabetes mellitus: a meta-analysis. Stem Cell Res Ther. 2021;12(1):273. doi: 10.1186/s13287-021-02342-5.
    1. Carlsson PO, Schwarcz E, Korsgren O, Le Blanc K. Preserved beta-cell function in type 1 diabetes by mesenchymal stromal cells. Diabetes. 2015;64(2):587–592. doi: 10.2337/db14-0656.
    1. Hu J, Yu X, Wang Z, Wang F, Wang L, Gao H, et al. Long term effects of the implantation of Wharton's jelly-derived mesenchymal stem cells from the umbilical cord for newly-onset type 1 diabetes mellitus. Endocr J. 2013;60(3):347–357. doi: 10.1507/endocrj.EJ12-0343.
    1. El-Badawy A, El-Badri N. Clinical efficacy of stem cell therapy for diabetes mellitus: a meta-analysis. PLoS ONE. 2016;11(4):e0151938. doi: 10.1371/journal.pone.0151938.
    1. Costa LA, Eiro N, Fraile M, Gonzalez LO, Saa J, Garcia-Portabella P, et al. Functional heterogeneity of mesenchymal stem cells from natural niches to culture conditions: implications for further clinical uses. Cell Mol Life Sci. 2021;78(2):447–467. doi: 10.1007/s00018-020-03600-0.
    1. Dunn CM, Kameishi S, Grainger DW, Okano T. Strategies to address mesenchymal stem/stromal cell heterogeneity in immunomodulatory profiles to improve cell-based therapies. Acta Biomater. 2021;133:114–125. doi: 10.1016/j.actbio.2021.03.069.
    1. Menard C, Dulong J, Roulois D, Hebraud B, Verdiere L, Pangault C, et al. Integrated transcriptomic, phenotypic, and functional study reveals tissue-specific immune properties of mesenchymal stromal cells. Stem Cells (Dayton, Ohio) 2020;38(1):146–159. doi: 10.1002/stem.3077.
    1. Shen WC, Lai YC, Li LH, Liao K, Lai HC, Kao SY, et al. Methylation and PTEN activation in dental pulp mesenchymal stem cells promotes osteogenesis and reduces oncogenesis. Nat Commun. 2019;10(1):2226. doi: 10.1038/s41467-019-10197-x.
    1. Li J, Xu SQ, Zhao YM, Yu S, Ge LH, Xu BH. Comparison of the biological characteristics of human mesenchymal stem cells derived from exfoliated deciduous teeth, bone marrow, gingival tissue, and umbilical cord. Mol Med Rep. 2018;18(6):4969–4977.
    1. Du WJ, Chi Y, Yang ZX, Li ZJ, Cui JJ, Song BQ, et al. Heterogeneity of proangiogenic features in mesenchymal stem cells derived from bone marrow, adipose tissue, umbilical cord, and placenta. Stem Cell Res Ther. 2016;7(1):163. doi: 10.1186/s13287-016-0418-9.
    1. Zhou T, Yuan Z, Weng J, Pei D, Du X, He C, et al. Challenges and advances in clinical applications of mesenchymal stromal cells. J Hematol Oncol. 2021;14(1):24. doi: 10.1186/s13045-021-01037-x.
    1. Xie Y, Liu W, Liu S, Wang L, Mu D, Cui Y, et al. The quality evaluation system establishment of mesenchymal stromal cells for cell-based therapy products. Stem Cell Res Ther. 2020;11(1):176. doi: 10.1186/s13287-020-01696-6.
    1. Han YF, Tao R, Sun TJ, Chai JK, Xu G, Liu J. Optimization of human umbilical cord mesenchymal stem cell isolation and culture methods. Cytotechnology. 2013;65(5):819–827. doi: 10.1007/s10616-012-9528-0.
    1. Liu W, Xie Y, Gao T, Huang F, Wang L, Ding L, et al. Reflection and observation: cell-based screening failing to detect HBV in HUMSCs derived from HBV-infected mothers underscores the importance of more stringent donor eligibility to reduce risk of transmission of infectious diseases for stem cell-based medical products. Stem Cell Res Ther. 2018;9(1):177. doi: 10.1186/s13287-018-0920-3.
    1. Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 2006;8(4):315–317. doi: 10.1080/14653240600855905.
    1. Lv W, Graves DT, He L, Shi Y, Deng X, Zhao Y, et al. Depletion of the diabetic gut microbiota resistance enhances stem cells therapy in type 1 diabetes mellitus. Theranostics. 2020;10(14):6500–6516. doi: 10.7150/thno.44113.
    1. Ezquer F, Ezquer M, Contador D, Ricca M, Simon V, Conget P. The antidiabetic effect of mesenchymal stem cells is unrelated to their transdifferentiation potential but to their capability to restore Th1/Th2 balance and to modify the pancreatic microenvironment. Stem Cells (Dayton, Ohio) 2012;30(8):1664–1674. doi: 10.1002/stem.1132.
    1. Bassi EJ, Moraes-Vieira PM, Moreira-Sa CS, Almeida DC, Vieira LM, Cunha CS, et al. Immune regulatory properties of allogeneic adipose-derived mesenchymal stem cells in the treatment of experimental autoimmune diabetes. Diabetes. 2012;61(10):2534–2545. doi: 10.2337/db11-0844.
    1. Shigemoto-Kuroda T, Oh JY, Kim DK, Jeong HJ, Park SY, Lee HJ, et al. MSC-derived extracellular vesicles attenuate immune responses in two autoimmune murine models: type 1 diabetes and uveoretinitis. Stem Cell Rep. 2017;8(5):1214–1225. doi: 10.1016/j.stemcr.2017.04.008.
    1. Honkanen J, Nieminen JK, Gao R, Luopajarvi K, Salo HM, Ilonen J, et al. IL-17 immunity in human type 1 diabetes. J Immunol (Baltimore, Md: 1950) 2010;185(3):1959–1967. doi: 10.4049/jimmunol.1000788.
    1. Emamaullee JA, Davis J, Merani S, Toso C, Elliott JF, Thiesen A, et al. Inhibition of Th17 cells regulates autoimmune diabetes in NOD mice. Diabetes. 2009;58(6):1302–1311. doi: 10.2337/db08-1113.
    1. Soliman H, Theret M, Scott W, Hill L, Underhill TM, Hinz B, et al. Multipotent stromal cells: one name, multiple identities. Cell Stem Cell. 2021;28(10):1690–1707. doi: 10.1016/j.stem.2021.09.001.
    1. Madec AM, Mallone R, Afonso G, Abou Mrad E, Mesnier A, Eljaafari A, et al. Mesenchymal stem cells protect NOD mice from diabetes by inducing regulatory T cells. Diabetologia. 2009;52(7):1391–1399. doi: 10.1007/s00125-009-1374-z.
    1. Zhang W, Zhou L, Dang J, Zhang X, Wang J, Chen Y, et al. Human gingiva-derived mesenchymal stem cells ameliorate streptozoticin-induced T1DM in mice via suppression of T effector cells and up-regulating Treg subsets. Sci Rep. 2017;7(1):15249. doi: 10.1038/s41598-017-14979-5.
    1. Araujo DB, Dantas JR, Silva KR, Souto DL, Pereira MFC, Moreira JP, et al. Allogenic adipose tissue-derived stromal/stem cells and vitamin D supplementation in patients with recent-onset type 1 diabetes mellitus: a 3-month follow-up pilot study. Front Immunol. 2020;11:993. doi: 10.3389/fimmu.2020.00993.
    1. Song M, Lee JH, Bae J, Bu Y, Kim EC. Human dental pulp stem cells are more effective than human bone marrow-derived mesenchymal stem cells in cerebral ischemic injury. Cell Transpl. 2017;26:1001–1016. doi: 10.3727/096368916X694391.
    1. Contentin R, Demoor M, Concari M, Desance M, Audigie F, Branly T, et al. Comparison of the chondrogenic potential of mesenchymal stem cells derived from bone marrow and umbilical cord blood intended for cartilage tissue engineering. Stem Cell Rev Rep. 2020;16(1):126–143. doi: 10.1007/s12015-019-09914-2.
    1. Shang F, Liu S, Ming L, Tian R, Jin F, Ding Y, et al. Human umbilical cord MSCs as new cell sources for promoting periodontal regeneration in inflammatory periodontal defect. Theranostics. 2017;7(18):4370–4382. doi: 10.7150/thno.19888.
    1. Mebarki M, Abadie C, Larghero J, Cras A. Human umbilical cord-derived mesenchymal stem/stromal cells: a promising candidate for the development of advanced therapy medicinal products. Stem Cell Res Ther. 2021;12(1):152. doi: 10.1186/s13287-021-02222-y.
    1. Baksh D, Yao R, Tuan RS. Comparison of proliferative and multilineage differentiation potential of human mesenchymal stem cells derived from umbilical cord and bone marrow. Stem Cells (Dayton, Ohio) 2007;25(6):1384–1392. doi: 10.1634/stemcells.2006-0709.
    1. Kern S, Eichler H, Stoeve J, Kluter H, Bieback K. Comparative analysis of mesenchymal stem cells from bone marrow, umbilical cord blood, or adipose tissue. Stem Cells (Dayton, Ohio) 2006;24(5):1294–1301. doi: 10.1634/stemcells.2005-0342.
    1. Huang Y, Li Q, Zhang K, Hu M, Wang Y, Du L, et al. Single cell transcriptomic analysis of human mesenchymal stem cells reveals limited heterogeneity. Cell Death Dis. 2019;10(5):368. doi: 10.1038/s41419-019-1583-4.
    1. Kassem DH, Kamal MM. Therapeutic efficacy of umbilical cord-derived stem cells for diabetes mellitus: a meta-analysis study. Stem Cell Res Ther. 2020;11(1):484. doi: 10.1186/s13287-020-01996-x.
    1. Lu J, Shen SM, Ling Q, Wang B, Li LR, Zhang W, et al. One repeated transplantation of allogeneic umbilical cord mesenchymal stromal cells in type 1 diabetes: an open parallel controlled clinical study. Stem Cell Res Ther. 2021;12(1):340. doi: 10.1186/s13287-021-02417-3.
    1. Najar M, Raicevic G, Boufker HI, Fayyad-Kazan H, De Bruyn C, Meuleman N, et al. Adipose-tissue-derived and Wharton's jelly-derived mesenchymal stromal cells suppress lymphocyte responses by secreting leukemia inhibitory factor. Tissue Eng Part A. 2010;16(11):3537–3546. doi: 10.1089/ten.tea.2010.0159.
    1. Amable PR, Teixeira MV, Carias RB, Granjeiro JM, Borojevic R. Protein synthesis and secretion in human mesenchymal cells derived from bone marrow, adipose tissue and Wharton's jelly. Stem Cell Res Ther. 2014;5(2):53. doi: 10.1186/scrt442.
    1. Shao S, He F, Yang Y, Yuan G, Zhang M, Yu X. Th17 cells in type 1 diabetes. Cell Immunol. 2012;280(1):16–21. doi: 10.1016/j.cellimm.2012.11.001.
    1. Weber SE, Harbertson J, Godebu E, Mros GA, Padrick RC, Carson BD, et al. Adaptive islet-specific regulatory CD4 T cells control autoimmune diabetes and mediate the disappearance of pathogenic Th1 cells in vivo. J Immunol (Baltimore, Md: 1950) 2006;176(8):4730–4739. doi: 10.4049/jimmunol.176.8.4730.

Source: PubMed

3
구독하다