Skip to main content
Diabetes logoLink to Diabetes
. 2012 May 14;61(6):1355–1356. doi: 10.2337/db12-0355

Mesenchymal Stem Cells for the Treatment of Diabetes

Antonello Pileggi 1,
PMCID: PMC3357279  PMID: 22618774

The field of regenerative medicine is rapidly evolving, paving the way for novel therapeutic interventions through cellular therapies and tissue engineering approaches that are reshaping the biomedical field. The remarkable plasticity of different cell subsets obtained from human embryonic and adult tissues from disparate sources (including bone marrow, umbilical cord, amniotic fluid, placenta, and adipose tissue) has sparked research endeavors evaluating use of these cells for numerous conditions, including diabetes and its complications (1).

A readily accessible source for multipotent stem cells is the bone marrow, which comprises progenitors of hematopoietic, endothelial, and mesenchymal stem cells (MSCs). Unfractioned and fractioned bone marrow–derived stem cells have been used in experimental and clinical settings to improve diabetes and diabetes complications. Bone marrow–derived MSCs are stromal, nonhematopoietic cells generally obtained from iliac crest aspirates following enrichment based on their preferential adhesion on culture vessels in defined media. MSC characterization relies on expression of specific surface markers and on their ability to differentiate into fat, bone, and cartilage when exposed to appropriate culture conditions (2).

Recent clinical trials have demonstrated powerful immunomodulatory effects of the inoculum of MSCs to treat graft-versus-host disease (3,4), to improve allogeneic renal transplant outcomes using lower immunosuppressive regimens (5), and to reduce immune cell activation in patients with multiple sclerosis and amyotrophic lateral sclerosis (6). Autologous MSCs were shown to improve Crohn disease lesions refractory to other therapies (7,8) and were tested for treatment of ischemic hearts (9).

In the context of diabetes research, MSCs have been used to generate insulin-producing cells (10), counteract autoimmunity (11,12), enhance islet engraftment and survival (13,14), and to treat diabetic ulcers and limb ischemia (15). Also, MSC inoculum improved metabolic control in experimental models of type 2 diabetes (T2D) (16). Nonrandomized, pilot trials in T2D suggest a positive impact of bone marrow–derived mononuclear cells on metabolic control (i.e., reduction of insulin requirements and of A1C values) in the absence of adverse events following intra-arterial injection by selective cannulation of the pancreas vasculature (17,18). Unfortunately, because these studies are small and lack in-depth mechanistic analyses, it is yet unknown how MSCs exert their beneficial effects in T2D.

The interesting study by Si et al. (19) attempts to understand the effects of autologous MSC inoculum in a rat model of T2D (induced by high-fat diet for 2 weeks followed by a suboptimal dose of the β-cell toxin streptozotocin [STZ] to induce a hyperglycemic state). Autologous MSCs were administered either 1 or 3 weeks after STZ treatment. Improved metabolic control, measured by enhanced insulin secretion, amelioration of insulin sensitivity, and increased islet numbers in the pancreas, was observed in animals receiving MSCs particularly when MSCs were given early (7 days) after STZ treatment. Consistent with previous reports, the metabolic effects of MSC inoculum were short-lived (for a period of 4 weeks), and reinoculum provided an additional, comparable, and transient effect.

Clamp studies demonstrated significantly improved blood glucose metabolism and insulin sensitivity in animals receiving MSC therapy. A set of novel mechanistic data emerging from this study indicates that MSC therapy is associated with improved insulin sensitivity via increased signaling (insulin receptor substrate-1 [IRS-1] and Akt phosphorylation upon feeding, as well as translocation of GLUT-4 on cell membrane upon insulin administration) in the muscle, liver, and adipose tissue of animals receiving MSC inoculum, when compared with controls. Although many questions remain unanswered, these data shed new light on the effects of autologous MSC inoculum on insulin target tissues in this rodent model of T2D.

It is important to consider the potential confounding elements that can be introduced by the disease model used in this and other similar studies. In the model used by Si et al., it is prudent to be cautious because of the relatively short duration of diabetes before initiation of the intervention. This model may not fully reflect the physiopathology of the progressive development of human T2D.

Si et al. (19) present important data that highlight the importance of a cautious interpretation of the results of their T2D model. For instance, the positive impact of MSC treatment on metabolic function was more pronounced when administered early (7 days) than late (21 days) after induction of diabetes. STZ is a naturally occurring nitrosourea with various biological actions as well as induction of acute and chronic cellular injury on several tissues, including pancreatic β-cells, liver, and kidney. In the animals receiving labeled MSC inoculum “early” after STZ (but not in those in the “late” STZ group), MSCs accumulated in pancreatic islets and liver where they may have contributed to tissue repair or remodeling, thereby mitigating the injury induced by STZ and a high-fat diet and improving metabolic function. Preservation of β-cell mass was also observed in the “early” MSC group, an observation that did not appear to result from increased replication (assessed by Ki67 immunoreactivity), but rather from tissue repair and the cytoprotective properties of MSCs.

MSCs offer new opportunities in the treatment of diabetes, but they also raise many scientific questions that need to be addressed, particularly those related to safety and efficacy. These issues have obvious implications for the clinical application of MSC and other innovative cellular therapies.

Further in-depth mechanistic studies are needed to understand how MSCs affect metabolic function in T2D and to help overcome the transient results that have been observed in several studies. It remains to be determined whether the diabetic microenvironment and/or comorbidities alter the quality or efficacy of MSCs isolated from and/or after inoculum in patients with diabetes (20). Identity, stability, potency, and safety of cellular products are of paramount importance in order to obtain reproducible results and prevent undesirable side effects.

The increasing body of evidence on the potential therapeutic properties of MSCs justifies cautious optimism concerning development of effective cellular therapies for treatment of diabetes in the future.

ACKNOWLEDGMENTS

This work was supported by grants from the National Institutes of Health (5U19AI050864-10, U01DK089538, 5U42RR016603-08S1, 1DP2DK083096-01, 1R01EB008009-02, 5R01DK059993-06, 1 R21 DK076098-01, 1 U01 DK70460-02, 5R01DK25802-24, 5R01DK56953-05), the Juvenile Diabetes Research Foundation International (17-2010-5, 4-2008-811, 6-39017G1, 4-2004-361, 4-2000-947), The Leona M. and Harry B. Helmsley Charitable Trust, the University of Miami Interdisciplinary Research Development Initiative, the Diabetes Research Institute Foundation, and Converge Biotech. The funders had no role in the content, presentation, decision to publish, or preparation of the manuscript.

A.P. is a co-founder, member of the scientific advisory board, and stock option holder of Converge Biotech and NEVA Pharmaceuticals. There are no patents, products in development, or marketed products to declare. A.P. has performed research supported by PositiveID Corporation, Extended Delivery Pharmaceuticals, Pfizer, Advanced Technologies, and Regenerative Medicine. No other potential conflicts of interest relevant to this article were reported.

Footnotes

See accompanying original article, p. 1616.

REFERENCES

  • 1.Fotino C, Ricordi C, Lauriola V, Alejandro R, Pileggi A. Bone marrow-derived stem cell transplantation for the treatment of insulin-dependent diabetes. Rev Diabet Stud 2010;7:144–157 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Dominici M, Le Blanc K, Mueller I, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 2006;8:315–317 [DOI] [PubMed] [Google Scholar]
  • 3.Le Blanc K, Rasmusson I, Sundberg B, et al. Treatment of severe acute graft-versus-host disease with third party haploidentical mesenchymal stem cells. Lancet 2004;363:1439–1441 [DOI] [PubMed] [Google Scholar]
  • 4.Le Blanc K, Frassoni F, Ball L, et al. Developmental Committee of the European Group for Blood and Marrow Transplantation Mesenchymal stem cells for treatment of steroid-resistant, severe, acute graft-versus-host disease: a phase II study. Lancet 2008;371:1579–1586 [DOI] [PubMed] [Google Scholar]
  • 5.Tan J, Wu W, Xu X, et al. Induction therapy with autologous mesenchymal stem cells in living-related kidney transplants: a randomized controlled trial. JAMA 2012;307:1169–1177 [DOI] [PubMed] [Google Scholar]
  • 6.Karussis D, Karageorgiou C, Vaknin-Dembinsky A, et al. Safety and immunological effects of mesenchymal stem cell transplantation in patients with multiple sclerosis and amyotrophic lateral sclerosis. Arch Neurol 2010;67:1187–1194 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Ciccocioppo R, Bernardo ME, Sgarella A, et al. Autologous bone marrow-derived mesenchymal stromal cells in the treatment of fistulising Crohn’s disease. Gut 2011;60:788–798 [DOI] [PubMed] [Google Scholar]
  • 8.Duijvestein M, Vos AC, Roelofs H, et al. Autologous bone marrow-derived mesenchymal stromal cell treatment for refractory luminal Crohn’s disease: results of a phase I study. Gut 2010;59:1662–1669 [DOI] [PubMed] [Google Scholar]
  • 9.Hare JM, Traverse JH, Henry TD, et al. A randomized, double-blind, placebo-controlled, dose-escalation study of intravenous adult human mesenchymal stem cells (prochymal) after acute myocardial infarction. J Am Coll Cardiol 2009;54:2277–2286 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Karnieli O, Izhar-Prato Y, Bulvik S, Efrat S. Generation of insulin-producing cells from human bone marrow mesenchymal stem cells by genetic manipulation. Stem Cells 2007;25:2837–2844 [DOI] [PubMed] [Google Scholar]
  • 11.Madec AM, Mallone R, Afonso G, et al. Mesenchymal stem cells protect NOD mice from diabetes by inducing regulatory T cells. Diabetologia 2009;52:1391–1399 [DOI] [PubMed] [Google Scholar]
  • 12.Fiorina P, Jurewicz M, Augello A, et al. Immunomodulatory function of bone marrow-derived mesenchymal stem cells in experimental autoimmune type 1 diabetes. J Immunol 2009;183:993–1004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Ding Y, Xu D, Feng G, Bushell A, Muschel RJ, Wood KJ. Mesenchymal stem cells prevent the rejection of fully allogenic islet grafts by the immunosuppressive activity of matrix metalloproteinase-2 and -9. Diabetes 2009;58:1797–1806 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Berman DM, Willman MA, Han D, et al. Mesenchymal stem cells enhance allogeneic islet engraftment in nonhuman primates. Diabetes 2010;59:2558–2568 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Lu D, Chen B, Liang Z, et al. Comparison of bone marrow mesenchymal stem cells with bone marrow-derived mononuclear cells for treatment of diabetic critical limb ischemia and foot ulcer: a double-blind, randomized, controlled trial. Diabetes Res Clin Pract 2011;92:26–36 [DOI] [PubMed] [Google Scholar]
  • 16.Lee RH, Seo MJ, Reger RL, et al. Multipotent stromal cells from human marrow home to and promote repair of pancreatic islets and renal glomeruli in diabetic NOD/scid mice. Proc Natl Acad Sci U S A 2006;103:17438–17443 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Bhansali A, Upreti V, Khandelwal N, et al. Efficacy of autologous bone marrow-derived stem cell transplantation in patients with type 2 diabetes mellitus. Stem Cells Dev 2009;18:1407–1416 [DOI] [PubMed] [Google Scholar]
  • 18.Estrada EJ, Valacchi F, Nicora E, et al. Combined treatment of intrapancreatic autologous bone marrow stem cells and hyperbaric oxygen in type 2 diabetes mellitus. Cell Transplant 2008;17:1295–1304 [DOI] [PubMed] [Google Scholar]
  • 19.Si Y, Zhao Y, Hao H, et al. Infusion of mesenchymal stem cells ameliorates hyperglycemia in type 2 diabetic rats: identification of a novel role in improving insulin sensitivity. Diabetes 2012;61:1616–1625 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Jurewicz M, Yang S, Augello A, et al. Congenic mesenchymal stem cell therapy reverses hyperglycemia in experimental type 1 diabetes. Diabetes 2010;59:3139–3147 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Diabetes are provided here courtesy of American Diabetes Association

RESOURCES