Join Us | Latest Articles | Contact

Journal Home


Editorial Board


Archive


Submit to this journal


Current issue

International Journal of Stem cell Research and Therapy





DOI: 10.23937/2469-570X/1410010



The Role of Mesenchymal Stem Cells in Diabetes Mellitus

Negar Azarpira1*, Maryam Kaviani1 and Saeede Salehi2


1Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
2Diagnostic Laboratory and Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran


*Corresponding author: Negar Azarpira, Shiraz University of Medical Sciences, Shiraz, Iran, Tel: +9-71-36473954, Fax: +9-71-36473954, E-mail: negarazarpira@yahoo.com
Int J Stem Cell Res Ther, IJSCRT-2-010, (Volume 2, Issue 2), Research Article; ISSN: 2469-570X
Received: May 01, 2015 | Accepted: June 30, 2015 | Published: July 02, 2015
Citation: Azarpira N, Kaviani M, Salehi S (2015) The Role of Mesenchymal Stem Cells in Diabetes Mellitus. Int J Stem Cell Res Ther 2:010. 10.23937/2469-570X/1410010
Copyright: © 2015 Azarpira N, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.



Abstract

Diabetes mellitus is a disease characterized by progressive destruction of the beta cells in the pancreatic islets of Langerhans. The current primary treatment is changing the life style and insulin injection. However, this therapy cannot provide sustained physiological glycemic control. Pancreas or islet cell transplantation would be the preferred treatment options. However, the lack of donor tissue immunoincompatibility, cell rejection and using long term immunosuppression are the major barriers to their widespread use.

Mesenchymal stem cells (MSC) hold promising therapy in regenerative medicine. MSCs are residing many adult and fetal tissue such as in the bone marrow, adipose tissue, umbilical cord blood, and Wharton jelly of the umbilical cord. MSCs, with self-renewal capacity and transdifferentiation potential are an attractive unlimited cell source for treating diabetic patients. On the other hand, MSCs have well characterized immunomodulatory properties that can suppress inflammatory damage and immune-mediated rejection and by secreting paracrine factors and deposition of the extracellular matrix improve the engraftment of pancreatic islets in micro-environmental niche. Here, we review the properties of MSCs and some of the recent clinical studies using MSCs as a new therapeutic option in the treatment of diabetic patients.


Keywords

Diabetes, Insulin, Beta cell, Stem cell, Islet


Introduction

Diabetes has become one of the most common chronic diseases in the world. The prevalence of diabetes was globally estimated to be 4.4% in 2030 and the total number of patients will be increased to 366 million [1].

Type 2 diabetes mellitus (T2DM) comprises 90% of all causes of diabetes and is characterized by a combination of peripheral insulin resistance and progressive beta-cell dysfunction. The routine treatments for T2DM include insulin sensitizers with exogenous insulin supply, but these drugs temporarily ameliorate hyperglycemia, and ultimately progressive beta cell dysfunction happens [2].

Type 1 diabetes mellitus (T1DM) comprises 10% of all causes of diabetes and is characterized by T cell-mediated destruction of insulin-producing cells in the pancreatic islets [1] during childhood. This autoimmune process (insulitis) is mainly mediated by CD4+ cells, CD8+ T cells, as well as proinflammatory cytokines, such as interleukin (IL)-2 and tumor necrosis factor (TNF)-a [3]. The patients are usually treated with exogenous insulin therapy. Nevertheless, a good metabolic control is not feasible and episodes of hypoglycemia and hyperglycemia frequently happen. Tight glycemic control is very important in diabetic patients [4]. But unfortunately, even with improved insulin formulations, the use of new infusion systems as well as continuous glucose monitoring, keeping tight control in the normal range is not possible. Therefore, chronic small vessel complications, such as retinopathy, nephropathy, and neuropathy occur [4,5].

Whole pancreas transplantation is another option for treatment, but it is associated with several limitations, such as major surgery, shortage of donors and organ rejection after transplantation. Islet cell transplantation is an alternative non invasive procedure in which the insulin secreting cells have physiological responses to the blood glucose levels [6]. The Edmonton group in 2000 established the "Edmonton Protocol" and demonstrated sustained long-term insulin-independence [6,7]. The islet cells are isolated from cadaveric donors [7,8], and injected into the recipient's portal vein. Instant blood-mediated inflammatory reactions, alloimmune reaction to transplanted cells and diabetogenic effect of immunosuppressive drugs reduced the initial beta cell mass and many patients require repeated episodes of cell transplantation [7,9].

Due to these limitations, the implantation of stem cells from embryonic or adult sources may be another potential treatment for diabetes [10,11]. Stem cells are characterized by their potential of self-renewal and multilineage differentiation. Therefore, they represent an alternative and unlimited source for differentiation toward islet cells.


Cell sources for generation of insulin producing cells (IPCs)

Previous in vitro studies showed that different types of stem cells such as embryonic stem cells [ESCs] [12], induced pleuripotent stem cells (iPS) [13], and mesenchymal stem cells (MSCs) [14] had been successfully differentiated into insulin-producing cells.

For differentiation of cells toward insulin producing cells, it is essential to pay attention to the native process of islets generation; and, the embryonic stem cell (ESC) differentiation protocols are useful models for understanding the molecular mechanism [15-17]. In this way, after establishment of pancreatic endoderm, inhibition of Notch signaling leads to further endocrine differentiation and, subsequently, maturation of endocrine precursors toward specialized insulin-producing cells occurs [18].

Nuclear reprogramming of adult somatic tissue leads to generation of autologous and patient-specific induced pluripotent stem (iPS) cells [13,19]. Differentiation of the skin fibroblast derived iPS cells toward insulin-producing islet-like clusters has been investigated. This cluster showed C-peptide and insulin expression, but the level of C-peptide secretion was lower compared with adult human islets. It was proposed that this low efficacy is in relation to differentiation procedure [20]. The generation of glucose responsive and functional islets requires silencing of stemness programs as well as the induction of stage specific transcription factors [21].

Routine differentiation protocols directing ESCs or iPS toward islets needs a variety of expensive cytokines and inhibitors. Due to the significance of microRNAs [miR] in islet development, such as miR-375 and Mir-7, their overexpression promotes pancreatic endocrine differentiation [22-24].

Different sources of MSCs, such as the umbilical cord blood, bone marrow and human pancreatic islet, have been investigated for differentiation to β-cells [25-27]. Wharton' jelly MSCs can be an important source for pancreatic islets generation due to possession of stem cell properties and easy achievement without ethical problems compared with embryonic and bone marrow stem cells. Transformation of Wharton' jelly MSCs into the islet-like cell clusters by neuron-conditioned medium has been analyzed. These clusters demonstrated insulin release and functional stability in vivo [28]. The researchers transfected MSCs by Pdx1 mRNA and induced them to differentiate by soluble factors. Strong expression of β-cells specific genes, Pdx1, Ngn3, Nkx6, and insulin, and the production of C-peptide and insulin were observed. The results of this study showed that induction of cells after transfection exhibits a better differentiation outcome than induction alone [29].

Generation of pancreatic islets from pancreatic progenitor cells has been also reported [30]. There are controversial studies about the existence of pancreatic progenitor cells [31]. It is speculated that progenitor cells are located within the basement membrane of common pancreatic ducts and they have the potential to differentiate and secrete insulin [30,31]. The differentiation of pancreatic progenitor cells derived from human fetal tissues was done by co-culture with liver stromal cells, devoid of growth factors. The examination of produced cells revealed the expression of important markers and functionality of the cells [32].


Mesenchymal Stem Cell with Dual Magic Function

MSCs are multi-potent, self-renewing cells that are isolated from different tissues, such as the bone marrow, adipose tissue [33], amniotic fluid [34], and umbilical cord blood [35]. The cells have a great multiplication potency and can be expanded in culture for several passages without losing their properties [36]. The International Society for Cellular Therapy has introduced criteria for defining these cells. MSCs adhere to plastic in culture plates and express cell surface markers, such as CD105, CD73, and CD90 and do not express CD45, CD34, CD14 or CD11b, CD79a, or CD19 and HLADR [37,38]. MSCs have been characterized for their ability to differentiate toward osteoblasts, adipocytes, and chondrocytes [39]. However, MSCs have also differentiated into endodermal and ectodermal lineages, including neural cells [40], hepatocytes [41], and insulin-producing cells [42].

Besides their differentiation potential toward insulin producing cells, MSCs contributed to repair processes through the secretion of pro-angiogenic molecules and formation of new blood vessels [43]. MSCs have well known unique immunomodulatory properties on T cells, B cells, dendritic cells and NK cells. The cells affect the role of regulatory T cells and autoreactive T cells by secreting several regulatory cytokines, such as IFN-γ, TGF-β, IL-4 and IL-10 [44].

MSCs also have an anti-inflammatory effect which is important in maintaining peripheral tolerance [45]. The cells express intermediate levels of MHC class I molecules but not the MHC class II which is important for rejection. Therefore, cell implantation across MHC barriers is possible. After cellular injury, these cells are able to migrate and settle in the injured tissues after systemic intravenous delivery [46].

After intravenous injection and migration to damaged tissues local over-expression of chemokines such as VCAM-1, SDF, MCP-1, CX3CL1-CX3CR1 and CXCL12-CXCR4 has been documented [47,48]. It seems that transplanted MSCs change the tissue microenvironment that supports the survival of damaged cell and inhibit the immune responses that accelerate the regeneration of homing recipient cells [47,48].

According to their immunomodulatory capacity, MSCs have been tried in clinical trials in treatment of steroid-refractory acute GVHD [49,50] and autoimmune diseases, such as multiple sclerosis, systemic lupus erythematosus [51] Crohn's disease [52] and Diabetes type 1 [53]. The results of these human trial revealed that repeated infusion of MSCs is safe and effective for treatment of at least a percentage of patients [54].

The immunomodulatory properties and paracrine effect of MScs on regeneration of resident beta cells was confirmed in differerent animal studies [55-57]. In diabetic patients.

In clinical trial database (http://www.clinicaltrials.gov) by April 2014, 28 human trials using MSC for diabetic patients were documented. In 8 projects, the trial was conducted on type 2 diabetic patients and in 20 studies, MSCs was transplantated to T1DM patients. The efficacy of these cells was also evaluated on chronic diabetic complications, such as diabetic foot. The MSCs were derived from the umbilical cord, outologous bone marrow and Prochymal Commercial drug [58-65].

The results of published clinical trials on stem cell transplantation in both diabetes type 1 and type 2 is summarized in table 1.


click here
Table 1: Clinical trials of MSC therapy for diabetes View Table 1


In an ongoing study, 21 patients with T1DM received outologous umbilical MSC infusions through the pancreatic artery. They believed that MSCs may change the local environment that promotes beta cell regeneration. They found that outologous UCB infusion was safe but failed to preserve C-peptide for a long time [58].

In another project, Hu et al. evaluated the long-term effects of the Wharton's jelly-derived MSCs on 29 patients with newly onset T1DM. The authors found that this treatment can restore the function of islet cells over a longer time [59].

The efficacy of autologous bone marrow mononuclear cells in the treatment of type 2 diabetes mellitus was also evaluated by this group and suggested that the implantation of autologous bone marrow mononuclear cells was safe and effective [60].

Mesples and co-workers used co-transplantation of MSCs and outologous HSCs stem cells in three T1DM patients. The bone marrow derived mononuclear cells were directly injected into the patient's liver paranchyma. During a long term follow-up, a significant increase in pancreatic secretion of C-peptide was documented [61]. In another clinical trial, twenty adult patients with newly diagnosed type 1 diabetes received MSC treatment. Residual β-cell function was analyzed with C-peptide in response to a mixed-meal tolerance test (MMTT). They found that autologous MSC treatment can contribute to the disease progression and preserve islet cell function [62].

Kong and colleagues used the umbilical cord derived MSCs for 18 patients with type 2 diabetes mellitus. The cells were transfused intravenously. They concluded that the umbilical cord derived MSCs transfusion was safe and it effectively alleviated blood glucose, and the C-peptide level was increased [63]. Another study was performed to evaluate the effect of combined autologous skin fibroblasts on biodegradable collagen membrane (Coladerm) in combination with outologous bone marrow derived mesenchymal stem cells for treatment of chronic non-healing wound in diabetic patients. They found that the wound size decreased and the vascularity of the dermis increased [64]. Dave and colleagues conducted another study on 10 insulin dependent diabetic patients with co-infusion of in vitro autologous adipose tissue-derived MSC-differentiated insulin-secreting cells (ISC) with hematopoietic stem cells (HSC). The results were promising and they suggested that this combination therapy offered a better long term control of hyperglycemia in patients [65].

Overall, in T1DM, the authors claimed that MSC therapy was safe and promising tool to intervene in disease progression and preserve function of β-cell. In type 2 diabetic patients MSC therapy was well tolerated, and this strategy effectively alleviated blood glucose level. In diabetic patients with critical limb ischemia, MSC therapy, accelerated the wound healing processes and decreased the rate of lower limb amputation [58-72].

In conclusion, the combination of immunomodulatory activity and tissue regenerative potential of MSCs as well as their differentiation capacity to islet like cells has attracted significant scientific and clinical interest.


References
  1. Liao YH, Verchere CB, Warnock GL (2007) Adult stem or progenitor cells in treatment for type 1 diabetes: current progress. Can J Surg 50: 137-142.

  2. Hatziavramidis DT, Karatzas TM, Chrousos GP (2013) Pancreatic islet cell transplantation: an update. Ann Biomed Eng 41: 469-476.

  3. Atkinson MA, Maclaren NK (1994) The pathogenesis of insulin-dependent diabetes mellitus. N Engl J Med 331: 1428-1436.

  4. Silverstein J, Klingensmith G, Copeland K, Plotnick L, Kaufman F, et al. (2005) Care of children and adolescents with type 1 diabetes: a statement of the American Diabetes Association. Diabetes Care 28: 186-212.

  5. Parving HH, Hommel E, Mathiesen E, Skøtt P, Edsberg B, et al. (1988) Prevalence of microalbuminuria, arterial hypertension, retinopathy and neuropathy in patients with insulin dependent diabetes. Br Med J (Clin Res Ed) 296: 156-160.

  6. Powers AC (2008) Insulin therapy versus cell-based therapy for type 1 diabetes mellitus: what lies ahead? Nat Clin Pract Endocrinol Metab 4: 664-665.

  7. Shapiro AM, Lakey JR, Ryan EA, Korbutt GS, Toth E, et al. (2000) Islet transplantation in seven patients with type 1 diabetes mellitus using a glucocorticoid-free immunosuppressive regimen. N Engl J Med 343: 230-238.

  8. Azarpira N, Aghdai MH, Nikeghbalian S, Geramizadeh B, Darai M, et al. (2014) Human islet cell isolation: the initial step in an islet transplanting program in Shiraz, Southern Iran. Exp Clin Transplant 12: 139-142.

  9. Agarwal A, Brayman KL (2012) Update on islet cell transplantation for type 1 diabetes. Semin Intervent Radiol 29: 90-98.

  10. Godfrey KJ, Mathew B, Bulman JC, Shah O, Clement S, et al. (2012) Stem cell-based treatments for Type 1 diabetes mellitus: bone marrow, embryonic, hepatic, pancreatic and induced pluripotent stem cells. Diabet Med 29: 14-23.

  11. Domínguez-Bendala J, Ricordi C (2012) Present and future cell therapies for pancreatic beta cell replenishment. World J Gastroenterol 18: 6876-6884.

  12. Basford CL, Prentice KJ, Hardy AB, Sarangi F, Micallef SJ, et al. (2012) The functional and molecular characterisation of human embryonic stem cell-derived insulin-positive cells compared with adult pancreatic beta cells. Diabetologia 55: 358-371.

  13. Kunisada Y, Tsubooka-Yamazoe N, Shoji M, Hosoya M (2012) Small molecules induce efficient differentiation into insulin-producing cells from human induced pluripotent stem cells. Stem Cell Res 8: 274-284.

  14. Koblas T, Zacharovová K, Berková Z, Leontovic I, Dovolilová E, et al. (2009) In vivo differentiation of human umbilical cord blood-derived cells into insulin-producing beta cells. Folia Biol (Praha) 55: 224-232.

  15. Sakano D, Shiraki N, Kume S (2015) Pancreatic Differentiation from Murine Embryonic Stem Cells. Methods Mol Biol.

  16. Shaer A, Azarpira N, Vahdati A, Karimi MH, Shariati M (2015) Differentiation of human-induced pluripotent stem cells into insulin-producing clusters. Exp Clin Transplant 13: 68-75.

  17. Conrad E, Stein R, Hunter CS2 (2014) Revealing transcription factors during human pancreatic ß cell development. Trends Endocrinol Metab 25: 407-414.

  18. Saito M, Kaneda A, Shigeto H, Hanata N, Otokuni K, et al. (2015) Development of an optimized 5-stage protocol for the in vitro preparation of insulin-secreting cells from mouse ES cells. Cytotechnology .

  19. Hosoya M (2012) Preparation of pancreatic ß-cells from human iPS cells with small molecules. Islets 4: 249-252.

  20. Tateishi K, He J, Taranova O, Liang G, D'Alessio AC, et al. (2008) Generation of insulin-secreting islet-like clusters from human skin fibroblasts. J Biol Chem 283: 31601-31607.

  21. Winkle M, van den Berg A, Tayari M, Sietzema J, Terpstra M, et al. (2015) Long noncoding RNAs as a novel component of the Myc transcriptional network. FASEB J 29: 2338-2346.

  22. Lahmy R, Soleimani M, Sanati MH, Behmanesh M, Kouhkan F, et al. (2013) Pancreatic islet differentiation of human embryonic stem cells by microRNA overexpression. J Tissue Eng Regen Med .

  23. Shaer A, Azarpira N, Karimi MH (2014) Differentiation of human induced pluripotent stem cells into insulin-like cell clusters with miR-186 and miR-375 by using chemical transfection. Appl Biochem Biotechnol 174: 242-258.

  24. Wu H, Mahato RI (2014) Mesenchymal stem cell-based therapy for type 1 diabetes. Discov Med 17: 139-143.

  25. Zanini C, Bruno S, Mandili G, Baci D, Cerutti F, et al. (2011) Differentiation of mesenchymal stem cells derived from pancreatic islets and bone marrow into islet-like cell phenotype. PLoS One 6: e28175.

  26. Van Pham P, Thi-My Nguyen P, Thai-Quynh Nguyen A, Minh Pham V, Nguyen-Tu Bui A, et al. (2014) Improved differentiation of umbilical cord blood-derived mesenchymal stem cells into insulin-producing cells by PDX-1 mRNA transfection. Differentiation 87: 200-208.

  27. Seyedi F, Farsinejad A, Moshrefi M, Nematollahi-Mahani SN (2015) In vitro evaluation of different protocols for the induction of mesenchymal stem cells to insulin-producing cells. In Vitro Cell Dev Biol Anim .

  28. Chao KC, Chao KF, Fu YS, Liu SH (2008) Islet-like clusters derived from mesenchymal stem cells in Wharton's Jelly of the human umbilical cord for transplantation to control type 1 diabetes. PLoS One 3: e1451.

  29. Van Pham P, Thi-My Nguyen P, Thai-Quynh Nguyen A, Minh Pham V, Nguyen-Tu Bui A, et al. (2014) Improved differentiation of umbilical cord blood-derived mesenchymal stem cells into insulin-producing cells by PDX-1 mRNA transfection. Differentiation 87: 200-208.

  30. Bonner-Weir S, Toschi E, Inada A, Reitz P, Fonseca SY, et al. (2004) The pancreatic ductal epithelium serves as a potential pool of progenitor cells. Pediatr Diabetes 5 Suppl 2: 16-22.

  31. Bonner-Weir S, Sharma A (2006) Are there pancreatic progenitor cells from which new islets form after birth? Nat Clin Pract Endocrinol Metab 2: 240-241.

  32. Liang J, Ng KY, Cheng Q, Xia Y, Wang CC, et al. (2014) Human fetal liver stromal cell co-culture enhances the differentiation of pancreatic progenitor cells into islet-like cell clusters. Stem Cell Rev 10: 280-294.

  33. Zuk PA, Zhu M, Mizuno H, Huang J, Futrell JW, et al. (2001) Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Eng 7: 211-228.

  34. In 't Anker PS, Scherjon SA, Kleijburg-van der Keur C, Noort WA, Claas FH, et al. (2003) Amniotic fluid as a novel source of mesenchymal stem cells for therapeutic transplantation. Blood 102: 1548-1549.

  35. Reddi AS, Kuppasani K, Ende N (2010) Human umbilical cord blood as an emerging stem cell therapy for diabetes mellitus. Curr Stem Cell Res Ther 5: 356-361.

  36. Polak JM, Bishop AE (2006) Stem cells and tissue engineering: past, present, and future. Ann N Y Acad Sci 1068: 352-366.

  37. Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, et al. (2006) Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 8: 315-317.

  38. Shaer A, Azarpira N, Aghdaie MH, Esfandiari E4 (2014) Isolation and characterization of Human Mesenchymal Stromal Cells Derived from Placental Decidua Basalis; Umbilical cord Wharton's Jelly and Amniotic Membrane. Pak J Med Sci 30: 1022-1026.

  39. Muñoz-Elias G, Marcus AJ, Coyne TM, Woodbury D, Black IB (2004) Adult bone marrow stromal cells in the embryonic brain: engraftment, migration, differentiation, and long-term survival. J Neurosci 24: 4585-4595.

  40. Schwartz RE, Reyes M, Koodie L, Jiang Y, Blackstad M, et al. (2002) Multipotent adult progenitor cells from bone marrow differentiate into functional hepatocyte-like cells. J Clin Invest 109: 1291-1302.

  41. Tang DQ, Cao LZ, Burkhardt BR, Xia CQ, Litherland SA, et al. (2004) In vivo and in vitro characterization of insulin-producing cells obtained from murine bone marrow. Diabetes 53: 1721-1732.

  42. Chen L, Tredget EE, Wu PY, Wu Y (2008) Paracrine factors of mesenchymal stem cells recruit macrophages and endothelial lineage cells and enhance wound healing. PLoS One 3: e1886.

  43. Kode JA, Mukherjee S, Joglekar MV, Hardikar AA (2009) Mesenchymal stem cells: immunobiology and role in immunomodulation and tissue regeneration. Cytotherapy 11: 377-391.

  44. Ge W, Jiang J, Baroja ML, Arp J, Zassoko R, et al. (2009) Infusion of mesenchymal stem cells and rapamycin synergize to attenuate alloimmune responses and promote cardiac allograft tolerance. Am J Transplant 9: 1760-1772.

  45. Seppanen E, Roy E, Ellis R, Bou-Gharios G, Fisk NM, et al. (2013) Distant mesenchymal progenitors contribute to skin wound healing and produce collagen: evidence from a murine fetal microchimerism model. PLoS One 8: e62662.

  46. Hanley PJ (2015) Therapeutic mesenchymal stromal cells: where we are headed. Methods Mol Biol 1283: 1-11.

  47. Shi Y, Su J, Roberts AI, Shou P, Rabson AB, et al. (2012) How mesenchymal stem cells interact with tissue immune responses. Trends Immunol 33: 136-143.

  48. Tolar J, Le Blanc K, Keating A, Blazar BR (2010) Concise review: hitting the right spot with mesenchymal stromal cells. Stem Cells 28: 1446-1455.

  49. Le Blanc K, Frassoni F, Ball L, Locatelli F, Roelofs H, et al. (2008) Mesenchymal stem cells for treatment of steroid-resistant, severe, acute graft-versus-host disease: a phase II study. Lancet 371: 1579-1586.

  50. Battiwalla M, Hematti P (2009) Mesenchymal stem cells in hematopoietic stem cell transplantation. Cytotherapy 11: 503-515.

  51. Liang J, Zhang H, Hua B, Wang H, Lu L, et al. (2010) Allogenic mesenchymal stem cells transplantation in refractory systemic lupus erythematosus: a pilot clinical study. Ann Rheum Dis 69: 1423-1429.

  52. Giordano A, Galderisi U, Marino IR (2007) From the laboratory bench to the patient's bedside: an update on clinical trials with mesenchymal stem cells. J Cell Physiol 211: 27-35.

  53. Domínguez-Bendala J, Lanzoni G, Inverardi L, Ricordi C (2012) Concise review: mesenchymal stem cells for diabetes. Stem Cells Transl Med 1: 59-63.

  54. Aali E, Mirzamohammadi S, Ghaznavi H, Madjd Z, Larijani B, et al. (2014) A comparative study of mesenchymal stem cell transplantation with its paracrine effect on control of hyperglycemia in type 1 diabetic rats. J Diabetes Metab Disord 13: 76.

  55. Glenn JD, Whartenby KA1 (2014) Mesenchymal stem cells: Emerging mechanisms of immunomodulation and therapy. World J Stem Cells 6: 526-539.

  56. Rahavi H, Hashemi SM, Soleimani M, Mohammadi J, Tajik N1 (2015) Adipose tissue-derived mesenchymal stem cells exert in vitro immunomodulatory and beta cell protective functions in streptozotocin-induced diabetic mice model. J Diabetes Res 2015: 878535.

  57. Hao H, Liu J, Shen J, Zhao Y, Liu H, et al. (2013) Multiple intravenous infusions of bone marrow mesenchymal stem cells reverse hyperglycemia in experimental type 2 diabetes rats. Biochem Biophys Res Commun 436: 418-423.

  58. Haller MJ, Wasserfall CH, Hulme MA, Cintron M, Brusko TM, (2013) Autologous umbilical cord blood infusion followed by oral docosahexaenoic acid and vitamin D supplementation for C-peptide preservation in children with Type 1 diabetes. Biol Blood Marrow Transplant. 19:1126-1129.

  59. Hu J, Yu X, Wang Z, Wang F, Wang L, et al. (2013) 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. 60:347-357.

  60. Hu J, Li C, Wang L, Zhang X, Zhang M, et al. (2012) Long term effects of the implantation of autologous bone marrow mononuclear cells for type 2 diabetes mellitus. Endocr J 59: 1031-1039.

  61. Mesples A, Majeed N, Zhang Y, Hu X (2013) Early immunotherapy using autologous adult stem cells reversed the effect of anti-pancreatic islets in recently diagnosed type 1 diabetes mellitus: preliminary results. Med Sci Monit. 19:852-857.

  62. Carlsson PO, Schwarcz E, Korsgren O, Le Blanc K4 (2015) Preserved ß-cell function in type 1 diabetes by mesenchymal stromal cells. Diabetes 64: 587-592.

  63. Kong D, Zhuang X, Wang D, Qu H, Jiang Y, et al. (2014) Umbilical cord mesenchymal stem cell transfusion ameliorated hyperglycemia in patients with type 2 diabetes mellitus. Clin Lab 60: 1969-1976.

  64. Vojtassák J, Danisovic L, Kubes M, Bakos D, Jarábek L, et al. (2006) Autologous biograft and mesenchymal stem cells in treatment of the diabetic foot. Neuro Endocrinol Lett 27 Suppl 2: 134-137.

  65. Dave SD, Vanikar AV, Trivedi HL, Thakkar UG, Gopal SC, et al. (2015) Novel therapy for insulin-dependent diabetes mellitus: infusion of in vitro-generated insulin-secreting cells. Clin Exp Med 15: 41-45.

  66. Zhao Y, Jiang Z, Zhao T, Ye M, Hu C, et al. (2012) Reversal of type 1 diabetes via islet ß cell regeneration following immune modulation by cord blood-derived multipotent stem cells. BMC Med 10: 3.

  67. Couri CE, Oliveira MC, Stracieri AB, Moraes DA, Pieroni F, et al. (2009) C-peptide levels and insulin independence following autologous nonmyeloablative hematopoietic stem cell transplantation in newly diagnosed type 1 diabetes mellitus. JAMA 301:1573-1579.

  68. Bhansali A, Asokumar P, Walia R, Bhansali S, Gupta V, et al. (2014) Efficacy and safety of autologous bone marrow-derived stem cell transplantation in patients with type 2 diabetes mellitus: a randomized placebo-controlled study. Cell Transplant 23: 1075-1085.

  69. Bhansali A, Upreti V, Walia R, Gupta V, Bhansali S, et al. (2014) Efficacy and safety of autologous bone marrow derived hematopoietic stem cell transplantation in patients with type 2 DM: A 15 months follow-up study. Indian J Endocrinol Metab. 18: 838-845.

  70. Mohammadzadeh L, Samedanifard SH, Keshavarzi A, Alimoghaddam K, Larijani B, et al. (2013) Therapeutic outcomes of transplanting autologous granulocyte colony-stimulating factor-mobilised peripheral mononuclear cells in diabetic patients with critical limb ischaemia. Exp Clin Endocrinol Diabetes.12:48-53.

  71. Jiang R, Han Z, Zhuo G, Qu X, Li X, et al. (2011) Transplantation of placenta-derived mesenchymal stem cells in type 2 diabetes: a pilot study. Front Med 5: 94-100.

  72. Lu D, Chen B, Liang Z, Deng W, Jiang Y, et al. (2011) 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. 92: 26-36.

  73. Bhansali A, Upreti V, Khandelwal N, Marwaha N, Gupta V, et al. (2009) Efficacy of autologous bone marrow-derived stem cell transplantation in patients with type 2 diabetes mellitus. Stem Cells Dev 18: 1407-1416.

International Journal of Anesthetics and Anesthesiology (ISSN: 2377-4630)
International Journal of Blood Research and Disorders   (ISSN: 2469-5696)
International Journal of Brain Disorders and Treatment (ISSN: 2469-5866)
International Journal of Cancer and Clinical Research (ISSN: 2378-3419)
International Journal of Clinical Cardiology (ISSN: 2469-5696)
Journal of Clinical Gastroenterology and Treatment (ISSN: 2469-584X)
Clinical Medical Reviews and Case Reports (ISSN: 2378-3656)
Journal of Dermatology Research and Therapy (ISSN: 2469-5750)
International Journal of Diabetes and Clinical Research (ISSN: 2377-3634)
Journal of Family Medicine and Disease Prevention (ISSN: 2469-5793)
Journal of Genetics and Genome Research (ISSN: 2378-3648)
Journal of Geriatric Medicine and Gerontology (ISSN: 2469-5858)
International Journal of Immunology and Immunotherapy (ISSN: 2378-3672)
International Journal of Medical Nano Research (ISSN: 2378-3664)
International Journal of Neurology and Neurotherapy (ISSN: 2378-3001)
International Archives of Nursing and Health Care (ISSN: 2469-5823)
International Journal of Ophthalmology and Clinical Research (ISSN: 2378-346X)
International Journal of Oral and Dental Health (ISSN: 2469-5734)
International Journal of Pathology and Clinical Research (ISSN: 2469-5807)
International Journal of Pediatric Research (ISSN: 2469-5769)
International Journal of Respiratory and Pulmonary Medicine (ISSN: 2378-3516)
Journal of Rheumatic Diseases and Treatment (ISSN: 2469-5726)
International Journal of Sports and Exercise Medicine (ISSN: 2469-5718)
International Journal of Stem Cell Research & Therapy (ISSN: 2469-570X)
International Journal of Surgery Research and Practice (ISSN: 2378-3397)
Trauma Cases and Reviews (ISSN: 2469-5777)
International Archives of Urology and Complications (ISSN: 2469-5742)
International Journal of Virology and AIDS (ISSN: 2469-567X)
More Journals

Contact Us

ClinMed International Library | Science Resource Online LLC
3511 Silverside Road, Suite 105, Wilmington, DE 19810, USA
Email: contact@clinmedlib.org
 

Feedback

Get Email alerts
 
Creative Commons License
Open Access
by ClinMed International Library is licensed under a Creative Commons Attribution 4.0 International License based on a work at https://clinmedjournals.org/.
Copyright © 2017 ClinMed International Library. All Rights Reserved.