Aslı Melike Ekmekçi1, Mai Abusalim1, Oytun Erbaş1

1ERBAS Institute of Experimental Medicine, Illinois, USA & Gebze, Türkiye

Keywords: Beta cells, diabetes mellitus, stem cells, stem cell therapy, embryonic stem cell.

Abstract

Diabetes mellitus (DM) is a widespread metabolic disease characterized by the disruption of blood glucose regulation, primarily caused by dysfunctional pancreatic beta (β)-cells. For the repair of β-cells, alternative approaches such as embryonic stem cells, mesenchymal stem cells, and induced pluripotent stem cells (iPSCs) are on the agenda due to the limitations of factors such as donor deficiency in islet cell transplantation treatment. It is aimed to produce real β-cells with the contributions of stem cell-based clinical studies conducted in recent years. In this chapter, stem cell transplantation is considered an alternative stem cellbased therapy in diabetes for insulin independence through various means such as β-cell differentiation and β-cell repair. Current and traditional treatment methods applied in Type 1 diabetes and Type 2 diabetes are not sufficient to prevent the devastating damage of microvascular and macrovascular complications. For this reason, promising stem cell approaches have been discussed in DM as well as its complications. This chapter focuses on the curative potential of cells with excellent differentiation ability, such as embryonic, adult, and iPSCs, in DM and its complications, which despite the discovery of insulin remain fatal.

Introduction

Glucose in the blood is regulated by beta (β)-cells secreted by the pancreas. Insulin plays a crucial role as a primary regulator of homeostasis since no other hormone is capable of reducing blood glucose levels. Diabetes mellitus (DM) is characterized by β-cell function loss, which leads to elevated blood glucose levels. The insulin-releasing pancreatic β-cells are destroyed or rendered ineffective, leading to DM. It is a metabolic condition that has spread worldwide. According to projections, it is estimated to reach 552 million cases in 2030.[1] There are two primary types: type 1 diabetes mellitus (T1DM) and type 2 diabetes mellitus (T2DM). The pathophysiology of T2DM comprises the development of resistance in insulin target tissues followed by β-cell malfunction due to a mix of genetic and environmental factors, in contrast to T1DM, which is defined by β-cell death leading to autoimmune dysfunction.[2] Monogenic diabetes, a less frequent form of the disease, is caused by a particular gene mutation that affects pancreas development and β-cell function.[3]

The literature demonstrates the availability of a range of therapeutic strategies for the management of diabetes. The most popular techniques include diet restriction, oral antidiabetic drugs, and insulin.[4-6]

One promising treatment is the exchange of β-cells via transplantation of islets of Langerhans, yet unfortunately, the lack of donors is the primary cause of its underuse. For this approach, human pluripotent stem cells (PSCs), such as embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), are a crucial supply of β-cells. With further studies, we have come remarkably close to the original form. However, the challenges of producing a fully mature β-cell remain.[7] This has the potential to be a real cure for T1DM and possibly T2DM and MD. Islet cell transplantation (ICT) has been associated with less progression of microvascular complications such as diabetic nephropathy (DNP), diabetic neuropathy (DN), diabetic retinopathy (DR), and others.[8] In different research with a three-year follow-up, ICT was superior to intensive medical therapy in terms of improving hemoglobin A1c and slowing the progression of DR.[9] Despite its ability to save lives, insulin often does not halt the development of end-stage microvascular complications in patients, thus, patients still face diabetes fatal consequences despite the development of insulin. Stem cell-based alternative therapy has eventually become a candidate for high interest over injected insulin as an important approach for diabetes treatment. Nonetheless, further research is needed to overcome various clinical challenges, such as donor shortages, and to determine its feasibility.

THE RELATIONSHIP BETWEEN STEM CELLS AND DIABETES

Transplantation of insulin-producing cells has enabled stem cell repair of pancreatic β-cells. [10,11] Under the normal conditions and signaling, stem cells have the astounding ability to self-renew and differentiate into specialized cells such as lymphocytes, hepatocytes, leukocytes, erythrocytes, myocytes, nerve cells, and muscle cells.[12]

As cell sources, stem cells are typically classified as ESCs or adult stem cells (ASCs). While ASCs are rare stem cells found in almost all major organs that are referred to as multipotent cells due to their limited ability to differentiate, ESCs -also known as PSCs- on the other hand, are differentiated from the embryo’s inner cell mass and have the ability to differentiate into various germ cell line.[13]

Adult stem cells are commonly found in medical applications. For instance, for the successful treatment of leukemia and other hematological tumors, bone marrow transplantation employs hematopoietic stem cells (HSCs) from donor marrow. In a similar manner to HCSs, ASCs not only can multiply but also differentiate into various blood cells, whereas mesenchymal stem cells (MSCs) promote the formation of fat, bone, and cartilage.[14,15] In recent years, remarkable progress has been made in the generation of functional β-cells from human stem cell populations. This strategy describes the path that PSCs take during embryogenesis, from definitive endoderm formation to pancreatic endoderm, endocrine progenitors, and ultimately islets of Langerhans.

Ethical concerns make investigating the prospect of regenerating insulin-secreting cells problematic.[16-18]

Scientists are attempting to employ several types of stem cells to treat a wide range of medical ailments.[19] Despite these advances, more than 400 million people with diabetes worldwide continue to suffer from catastrophic consequences such as DNP, DN, and DR.[20]

Diabetes occurs when the pancreatic cells responsible for insulin secretion become dysfunctional or produce insufficient insulin, the body does not respond to the produced insulin, and glucose builds up in the blood. As a result of this inability to manage glucose, diabetes-related micro-, and macrovascular effects occur. Thirst, polyphagia, weight changes, polyuria, and blurred vision are common symptoms of diabetes. In advanced cases, hyperglycemia with ketoacidosis is likely to occur.[21-24]

PLURIPOTENT STEM CELLS AND DIABETES

Scientists highly value the pluripotent state of ESCs, and it is for that, that they are being studied for their use in a variety of medical conditions, including diabetes.[25] Through differentiation and established development, ESCs are viewed as a great source for the production of islet cells capable of producing insulin. Although challenging when considered, it is possible that ESCs might be made to differentiate into pancreatic islet cells, which then could be transplanted into the area of concern in diabetic patients, thus preventing β-cell deficiency. In the past, mouse ESCs (mESCs) have been used for this approach. Researchers have generated replicas from genetically altered and drug-selected mESCs that can secrete insulin. Following monitoring, these cells were implanted into diabetic mice and improved hyperglycemia.[26–31] Aside from mESCs, another group utilized human ESCs (hESCs) for the same purpose.[32,33]

Cells co-expressing pancreatic and duodenal homeobox 1 (PDX1) and NK6 homeobox protein 1 (NKX6.1) in the developing human embryo show multipotent pancreatic bud and stem progenitors that subsequently produce insulin-secreting β-cells. [34]

Key transcription factors (TFs) are highly expressed in pancreatic progenitor cells and β-cells involved in insulin secretion. Co-expression of PDX1 and NKX6.1 has been shown to be essential for the production of mono-hormonal, glucose-sensitive β-cells. [35,36]

Specifically, NKX6.1 is a crucial marker regulating β-cell maturation and functionality.[35,37] Researchers have reported varying degrees of success with regard to ESCs and islet generation. As a result, many issues have been encountered, including cell homogeneity, immaturity of differentiated cells, low numbers of cells that produce insulin, and inadequate insulin sensitivity to glucose.[30,32,33,38,39] On the other hand, as neither C-peptide nor intracellular insulin is produced after the cells are cultivated in an insulin-free medium, several research groups claim that these cells are not insulin-producing cells at all.[40-42]

The first cell line to be used for in vitro produce β-cells were ESC cells. A procedure has been created by one group to transform mESCs into definitive, completely pure, endodermal cell lines.[16] It demonstrated the production of pancreatic endocrine hormone-producing cells containing insulin and C-peptide.[43] As a result, they were able to produce insulin from these cells in the human islet interval, yet were unable to produce it in response to glucose. Later on, this response was achieved by a different group. Pluripotent stem cells have been proven to have drawbacks, including a significant risk of tumorigenesis, immunological rejection, and ethical controversies.[18,44-46] These considerations explain the reason why the clinical use of ESCs is still unclear. Numerous molecular similarities are shared between iPSCs and ESCs. Therefore, by obtaining specific iPSCs from diabetics, the ethical and immunological rejection concerns and moral questions associated with ESC transplantation have not emerged.[47-54] These findings might make iPSCs a promising choice for cellular replacement therapy in T1DM in the future.

STEM CELL TREATMENT FOR T2DM

Type 2 diabetes mellitus is characterized by insulin resistance and reduced insulin secretion. Treatment includes diet, oral antidiabetics, and the use of external insulin.[55-64] Patients with T2DM who regularly take insulin eventually acquire insulin resistance, and existing therapies do not completely solve this issue.[65] Although transplanting pancreatic islet cells is seen to be a viable strategy, obstacles like a paucity of donors and ethical concerns have limited its use. In order to increase the lowered insulin levels in patients, stem cells such BMSCs, ADSCs, ESCs, and iPSCs can develop into beta- and comparable cells capable of producing insulin.[45,66]

Patients with T2DM who received a combination of intrapancreatic bone marrow infusion and hyperbaric oxygen therapy experienced improvements in glycemic control and C-peptide levels as well as a reduction in their need for insulin.[67] After receiving a BMSCs injection, T2DM patients improved in the same way.[68]

In rats with high-fat diet-induced T2DM, BM-MSC transplantation activated insulin receptor substrate, and reduced hyperglycemia. It was discovered that glucose transporter type 4 (GLUT4) translocation and expression had increased.[69]

Mesenchymal stem cells have demonstrated therapeutic effects on islet cell recovery and glycemic control in animal models. Clinical practice has been affected by these findings. The literature contains clinical research on MSC therapy in T2DM patients.[70-78] Nevertheless, there is still a long way to go for a definitive and routine approach to stem cell-based treatment of T2DM.

Recent research has demonstrated that VEGF is crucial to the development of vascular damage in DR and has suggested that blocking VEGF is a useful strategy for managing the condition. The reduction of VEGF production by MSC injection in a hypoxic environment by the reductase enzyme inhibitor atorvastatin has been proven.[79-85] Moreover, studies indicate that BM-HSCs provide better visual activity.[86]

Epithelial progenitor cells (EPCs) generated from mouse BM-MSCs and human MSCs have been demonstrated in animal models to stimulate neovascularization and enhance DR.[87-89]

Patients with T1DM and T2DM may develop foot ulcers and require amputations as a result of DN, one of the most prevalent consequences of DM. When hyperglycemia rises over time, DN develops into a chronic condition.[90,91] Among the reasons linked to the occurrence of DN are dysregulated glucose levels, metabolic variables, oxidative stress, elevated glycolysis hemoglobin levels, and poor blood velocity due to free radical buildup.[91,92] Besides, prolonged elevated blood glucose levels also promote the creation of advanced glycation end products (AGEs), which, after binding to their receptors, start an inflammatory reaction and enhance oxidative stress, which further causes Schwann cells to deteriorate. Subsequently, any oxidation-mediated loss of function in these cells, which govern nerve regeneration as well as neuron insulation, increases DN in diabetes patients.[93-98]

Diabetic nephropathy, a microvascular complication of DM, is one of the most common causes of end-stage chronic kidney disease and is associated with high mortality.[99-101] Matrix molecule-producing podocytes in the glomerular basal membrane are damaged in DNP, resulting in proteinuria, fibrosis, and renal failure. Self-regeneration of damaged podocytes is limited, and the proteinuria condition worsens due to the negative effect on the glomerular barrier.[102]

Proteinuria, fibrosis, and dysfunction of proximal tubular epithelial cells (PTECs) together with increased tubulointerstitial inflammation are all signs of decreased renal function.[103] The negative features of PTECs, such as inflammation, are increased by prolonged hyperglycemia, AGEs, and glycated albumin.[104]

The renin-angiotensin system activation, synthesis of different growth factors, and excessive cytokine production are only a few of the several routes whereby AGEs are hypothesized to be implicated in the pathophysiology of DNP.[105] By preventing the production of pro-inflammatory cytokines, blocking inducible nitric oxide synthase, and encouraging parenchymal cell proliferation, MSCs can improve renal healing.[106,107] To simulate DNP characteristics, iPSCs were developed into podocytes in many studies.[108,109]

The paracrine action of renal trophic factors released by MSCs in DNP was the subject of one investigation. Animals with diabetes brought on by a high-fat diet and streptozotocin received MSCs. It was found that both therapies had ameliorative effects.[110] A significant decrease in blood glucose levels was observed in MSC-treated diabetic mice. Furthermore, albuminuria was reduced, and glomeruli were histologically normal in these animals. On the other hand, in diabetic mice without MSC treatment, glomerular enlargement was found to be present. Thus, MSC administration appeared to prevent the regeneration of beta-pancreatic islets and kidney damage in diabetic animals. According to the results of the study, MSC transplantation is recommended as a treatment for T1DM.[111] In addition to that, by reducing podocyte loss and promoting the release of bone morphogenetic protein-7, MSCs reduced fibrosis and glomerulosclerosis. They thereby contributed to the regeneration and protection of DNP.[112]

The injection of BM-MSC enhanced renal function and controlled the levels of insulin, heme oxygenase-1, AGEs, and glucose in the blood.[113] The results of the research show that stem cell-based treatments, such as MSCs, are successful in treating DNP, despite their limitations due to the consequences mentioned previously.

POTENTIAL OF STEM CELLS: THEIR IMPACT ON MACROVASCULAR AND BEYOND

Atherosclerosis is a macrovascular condition that is common in DM patients. Stroke, myocardial infarction, and vascular disease are among the risks that have been linked to persistently elevated blood sugar levels.[114,115] Depletion EPCs and the presence of cells like CD133 and CD34 are reliable indicators of arterial disease. Moreover, reduced EPC numbers have been identified as a potential new indicator of peripheral artery disease in DM.[116–118]

Vascular stem cells, which may identify EPCs, are being researched as a potential therapy for the macrovascular problems of diabetes. In one study, it was demonstrated that vascular progenitor cells developed from human vascular smooth muscle cells into vascular networks.[119] In vivo testing of EPCs’ capacity to create vascular networks was successful. The same CD133+ subset from which mesenchymal progenitor cells (MPCs) are produced may also be a candidate for this vascular job.[120,121] Intravenous injection of MPCs slowed cardiac remodeling and enhanced myocardial function in a diabetic animal investigation employing a cardiomyopathy model, with a substantial increase in matrix metalloproteinase (MMP)-2 activity and a decrease in MMP-9.[122]

Considering in terms of long-term implications in DM, chronic hyperglycemia is known to cause endothelial dysfunction, subsequently causing issues including vascular network damage in the target organs. The ability of progenitor cells from diabetic animals to restore vascular homeostasis has been demonstrated in various experiments.[123,124] This finding implies that the number of stem cells decreases with the formation of a deficit of major stem cells in diabetes. The use of these formerly mentioned two stem cells to correct vascular dysfunction and restore vascular function still requires further research before a clear prescription can be made. Nonetheless, the use of stem cells to treat macrovascular problems appears promising.

In conclusion, diabetes is a metabolic condition that is widespread across the world. Due to damage to the pancreas’ β-cells, it is characterized by insulin loss and impaired insulin sensitivity. Diabetes and its consequences continue to endanger human life despite the discovery of insulin. Although ICT has been tested by researchers as an alternate therapy, the lack of donors still poses problems in practice. Furthermore, the first stem cells employed in the stem cell strategy for diabetes were ESCs. Yet, iPSCs have emerged as a substitute due to issues including tumor risk as well as ethical questions. Mesenchymal stem cells and BM-HSCs have also been alternative sources for β-cells. Induced pluripotent stem cells regulate glucose by developing into beta-cell-like cells, according to animal model research. We covered the microvascular and macrovascular effects of diabetes in this chapter, as well as prospective therapeutic strategies using the current stem cell paradigm. Mesenchymal and HSCs have been demonstrated to aid in retinal healing in DR by differentiating into ocular cells. Similarly, stem cell applications for DNP, DR, and atherogenic illnesses brought on by endothelial dysfunction caused by diabetes are being studied. Given intercellular communication, heterogeneity, tumor risk, and ethical considerations, cells with this remarkable capacity for differentiation are likely candidates to be used in the development of future standard operating procedures to treat diabetes and its complications by substituting insulin, which has no lasting effects.

Cite this article as: Ekmekçi AM, Abusalim M, Erbaş O. Stem Cell Therapy for Diabetes Treatment. JEB Med Sci 2024;5(1):60-68.

Conflict of Interest

The authors declared no conflicts of interest with respect to the authorship and/or publication of this article.

Financial Disclosure

The authors received no financial support for the research and/or authorship of this article.

References

  1. Whiting DR, Guariguata L, Weil C, Shaw J. IDF diabetes atlas: global estimates of the prevalence of diabetes for 2011 and 2030. Diabetes Res Clin Pract. 2011 Dec;94:311-21.
  2. Cnop M, Welsh N, Jonas JC, Jörns A, Lenzen S, Eizirik DL. Mechanisms of pancreatic beta-cell death in type 1 and type 2 diabetes: many differences, few similarities. Diabetes. 2005 Dec;54 Suppl 2:S97-107.
  3. Murphy R, Ellard S, Hattersley AT. Clinical implications of a molecular genetic classification of monogenic beta-cell diabetes. Nat Clin Pract Endocrinol Metab. 2008 Apr;4:200-13.
  4. Yamada S, Kabeya Y, Noto H. Dietary Approaches for Japanese Patients with Diabetes: A Systematic Review. Nutrients. 2018 Aug 13;10:1080.
  5. Phung OJ, Sood NA, Sill BE, Coleman CI. Oral anti-diabetic drugs for the prevention of Type 2 diabetes. Diabet Med. 2011 Aug;28:948-64.
  6. Lee SH, Yoon KH. A Century of Progress in Diabetes Care with Insulin: A History of Innovations and Foundation for the Future. Diabetes Metab J. 2021 Sep;45:629-40.
  7. Bourgeois S, Sawatani T, Van Mulders A, De Leu N, Heremans Y, Heimberg H, et al. Towards a Functional Cure for Diabetes Using Stem Cell-Derived Beta Cells: Are We There Yet? Cells. 2021 Jan 19;10:191.
  8. Thompson DM, Meloche M, Ao Z, Paty B, Keown P, Shapiro RJ, et al. Reduced progression of diabetic microvascular complications with islet cell transplantation compared with intensive medical therapy. Transplantation. 2011 Feb 15;91:373-8.
  9. Warnock GL, Thompson DM, Meloche RM, Shapiro RJ, Ao Z, Keown P, et al. A multi-year analysis of islet transplantation compared with intensive medical therapy on progression of complications in type 1 diabetes. Transplantation. 2008 Dec 27;86:1762-6.
  10. McCall MD, Toso C, Baetge EE, Shapiro AM. Are stem cells a cure for diabetes? Clin Sci (Lond). 2009 Oct 12;118:87-97.
  11. Soria B, Bedoya FJ, Tejedo JR, Hmadcha A, Ruiz-Salmerón R, Lim S, et al. Cell therapy for diabetes mellitus: an opportunity for stem cells? Cells Tissues Organs. 2008;188:70-7.
  12. Old Protein, New Medicine - Brain-Derived Neurotrophic Factor [Working Title] [Internet]. Biochemistry. IntechOpen; 2024. Available from: http://dx.doi. org/10.5772/intechopen.111201
  13. Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ, Marshall VS, et al. Embryonic stem cell lines derived from human blastocysts. Science. 1998 Nov 6;282:1145-7.
  14. Krause DS, Theise ND, Collector MI, Henegariu O, Hwang S, Gardner R, et al. Multi-organ, multi-lineage engraftment by a single bone marrow-derived stem cell. Cell. 2001 May 4;105:369-77.
  15. Jiang Y, Jahagirdar BN, Reinhardt RL, Schwartz RE, Keene CD, Ortiz-Gonzalez XR, et al. Pluripotency of mesenchymal stem cells derived from adult marrow. Nature. 2002 Jul 4;418:41-9.
  16. D'Amour KA, Agulnick AD, Eliazer S, Kelly OG, Kroon E, Baetge EE. Efficient differentiation of human embryonic stem cells to definitive endoderm. Nat Biotechnol. 2005 Dec;23:1534-41.
  17. Rezania A, Bruin JE, Riedel MJ, Mojibian M, Asadi A, Xu J, et al. Maturation of human embryonic stem cell-derived pancreatic progenitors into functional islets capable of treating pre-existing diabetes in mice. Diabetes. 2012 Aug;61:2016-29.
  18. Zhu S, Russ HA, Wang X, Zhang M, Ma T, Xu T, et al. Human pancreatic beta-like cells converted from fibroblasts. Nat Commun. 2016 Jan 6;7:10080.
  19. Liu X, Wang Y, Li Y, Pei X. Research status and prospect of stem cells in the treatment of diabetes mellitus. Sci China Life Sci. 2013 Apr;56:306-12.
  20. Diabetes Control and Complications Trial Research Group; Nathan DM, Genuth S, Lachin J, Cleary P, Crofford O, Davis M, et al. The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus. N Engl J Med. 1993 Sep 30;329:977-86.
  21. Bratanova-Tochkova TK, Cheng H, Daniel S, Gunawardana S, Liu YJ, Mulvaney-Musa J, et al. Triggering and augmentation mechanisms, granule pools, and biphasic insulin secretion. Diabetes. 2002 Feb;51 Suppl 1:S83-90.
  22. American Diabetes Association. Diagnosis and classification of diabetes mellitus. Diabetes Care. 2014 Jan;37 Suppl 1:S81-90.
  23. Li M, Ikehara S. Stem cell treatment for type 1 diabetes. Front Cell Dev Biol. 2014 Mar 20;2:9.
  24. Chhabra P, Brayman KL. Stem cell therapy to cure type 1 diabetes: from hype to hope. Stem Cells Transl Med. 2013 May;2:328-36.
  25. Trounson A. A rapidly evolving revolution in stem cell biology and medicine. Reprod Biomed Online. 2013 Dec;27:756-64.
  26. Soria B, Roche E, Berná G, León-Quinto T, Reig JA, Martín F. Insulin-secreting cells derived from embryonic stem cells normalize glycemia in streptozotocin-induced diabetic mice. Diabetes. 2000 Feb;49:157-62.
  27. Blyszczuk P, Asbrand C, Rozzo A, Kania G, St-Onge L, Rupnik M, et al. Embryonic stem cells differentiate into insulin-producing cells without selection of nestin-expressing cells. Int J Dev Biol. 2004 Dec;48:1095-104.
  28. Hori Y, Rulifson IC, Tsai BC, Heit JJ, Cahoy JD, Kim SK. Growth inhibitors promote differentiation of insulinproducing tissue from embryonic stem cells. Proc Natl Acad Sci U S A. 2002 Dec 10;99:16105-10.
  29. Kahan BW, Jacobson LM, Hullett DA, Ochoada JM, Oberley TD, Lang KM, et al. Pancreatic precursors and differentiated islet cell types from murine embryonic stem cells: an in vitro model to study islet differentiation. Diabetes. 2003 Aug;52:2016-24.
  30. Lumelsky N, Blondel O, Laeng P, Velasco I, Ravin R, McKay R. Differentiation of embryonic stem cells to insulin-secreting structures similar to pancreatic islets. Science. 2001 May 18;292:1389-94.
  31. León-Quinto T, Jones J, Skoudy A, Burcin M, Soria B. In vitro directed differentiation of mouse embryonic stem cells into insulin-producing cells. Diabetologia. 2004 Aug;47:1442-51.
  32. Assady S, Maor G, Amit M, Itskovitz-Eldor J, Skorecki KL, Tzukerman M. Insulin production by human embryonic stem cells. Diabetes. 2001 Aug;50:1691-7.
  33. Segev H, Fishman B, Ziskind A, Shulman M, Itskovitz-Eldor J. Differentiation of human embryonic stem cells into insulin-producing clusters. Stem Cells. 2004;22:265-74.
  34. Al-Khawaga S, Memon B, Butler AE, Taheri S, Abou-Samra AB, Abdelalim EM. Pathways governing development of stem cell-derived pancreatic β cells: lessons from embryogenesis. Biol Rev Camb Philos Soc. 2018 Feb;93:364-89.
  35. Rezania A, Bruin JE, Xu J, Narayan K, Fox JK, O'Neil JJ, et al. Enrichment of human embryonic stem cell-derived NKX6.1-expressing pancreatic progenitor cells accelerates the maturation of insulin-secreting cells in vivo. Stem Cells. 2013 Nov;31:2432-42.
  36. Jennings RE, Berry AA, Kirkwood-Wilson R, Roberts NA, Hearn T, Salisbury RJ, et al. Development of the human pancreas from foregut to endocrine commitment. Diabetes. 2013 Oct;62:3514-22.
  37. Taylor BL, Liu FF, Sander M. Nkx6.1 is essential for maintaining the functional state of pancreatic beta cells. Cell Rep. 2013 Sep 26;4:1262-75.
  38. Hori Y, Gu X, Xie X, Kim SK. Differentiation of insulin-producing cells from human neural progenitor cells. PLoS Med. 2005 Apr;2:e103.
  39. Miyazaki S, Yamato E, Miyazaki J. Regulated expression of pdx-1 promotes in vitro differentiation of insulin-producing cells from embryonic stem cells. Diabetes. 2004 Apr;53:1030-7.
  40. Hansson M, Tonning A, Frandsen U, Petri A, Rajagopal J, Englund MC, et al. Artifactual insulin release from differentiated embryonic stem cells. Diabetes. 2004 Oct;53:2603-9.
  41. Rajagopal J, Anderson WJ, Kume S, Martinez OI, Melton DA. Insulin staining of ES cell progeny from insulin uptake. Science. 2003 Jan 17;299:363.
  42. Sipione S, Eshpeter A, Lyon JG, Korbutt GS, Bleackley RC. Insulin expressing cells from differentiated embryonic stem cells are not beta cells. Diabetologia. 2004 Mar;47:499-508.
  43. D'Amour KA, Bang AG, Eliazer S, Kelly OG, Agulnick AD, Smart NG, et al. Production of pancreatic hormone-expressing endocrine cells from human embryonic stem cells. Nat Biotechnol. 2006 Nov;24:1392-401.
  44. Kroon E, Martinson LA, Kadoya K, Bang AG, Kelly OG, Eliazer S, et al. Pancreatic endoderm derived from human embryonic stem cells generates glucose-responsive insulin-secreting cells in vivo. Nat Biotechnol. 2008 Apr;26:443-52.
  45. Pagliuca FW, Millman JR, Gürtler M, Segel M, Van Dervort A, Ryu JH, et al. Generation of functional human pancreatic β cells in vitro. Cell. 2014 Oct 9;159:428-39.
  46. Kim JB, Zaehres H, Wu G, Gentile L, Ko K, Sebastiano V, et al. Pluripotent stem cells induced from adult neural stem cells by reprogramming with two factors. Nature. 2008 Jul 31;454:646-50.
  47. Chandra V, G S, Phadnis S, Nair PD, Bhonde RR. Generation of pancreatic hormone-expressing islet-like cell aggregates from murine adipose tissue-derived stem cells. Stem Cells. 2009 Aug;27:1941-53.
  48. Lilly MA, Davis MF, Fabie JE, Terhune EB, Gallicano GI. Current stem cell based therapies in diabetes. Am J Stem Cells. 2016 Oct 20;5:87-98.
  49. Godfrey KJ, Mathew B, Bulman JC, Shah O, Clement S, Gallicano GI. Stem cell-based treatments for Type 1 diabetes mellitus: bone marrow, embryonic, hepatic, pancreatic and induced pluripotent stem cells. Diabet Med. 2012 Jan;29:14-23.
  50. Wagner RT, Lewis J, Cooney A, Chan L. Stem cell approaches for the treatment of type 1 diabetes mellitus. Transl Res. 2010 Sep;156:169-79.
  51. Hussain MA, Theise ND. Stem-cell therapy for diabetes mellitus. Lancet. 2004 Jul 10-16;364:203-5.
  52. Blyszczuk P, Czyz J, Kania G, Wagner M, Roll U, St-Onge L, et al. Expression of Pax4 in embryonic stem cells promotes differentiation of nestin-positive progenitor and insulin-producing cells. Proc Natl Acad Sci U S A. 2003 Feb 4;100:998-1003.
  53. Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007 Nov 30;131:861-72.
  54. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006 Aug 25;126:663-76.
  55. Alipio Z, Liao W, Roemer EJ, Waner M, Fink LM, Ward DC, et al. Reversal of hyperglycemia in diabetic mouse models using induced-pluripotent stem (iPS)-derived pancreatic beta-like cells. Proc Natl Acad Sci U S A. 2010 Jul 27;107:13426-31.
  56. Granger A, Kushner JA. Cellular origins of beta-cell regeneration: a legacy view of historical controversies. J Intern Med. 2009 Oct;266:325-38.
  57. Weir GC, Cavelti-Weder C, Bonner-Weir S. Stem cell approaches for diabetes: towards beta cell replacement. Genome Med. 2011 Sep 27;3:61.
  58. Cao LZ, Tang DQ, Horb ME, Li SW, Yang LJ. High glucose is necessary for complete maturation of Pdx1-VP16-expressing hepatic cells into functional insulin-producing cells. Diabetes. 2004 Dec;53:3168-78.
  59. Fujimoto K, Polonsky KS. Pdx1 and other factors that regulate pancreatic beta-cell survival. Diabetes Obes Metab. 2009 Nov;11 Suppl 4:30-7.
  60. Zalzman M, Gupta S, Giri RK, Berkovich I, Sappal BS, Karnieli O, et al. Reversal of hyperglycemia in mice by using human expandable insulin-producing cells differentiated from fetal liver progenitor cells. Proc Natl Acad Sci U S A. 2003 Jun 10;100:7253-8.
  61. Dang LT, Kim NP, Truong KD. Mesenchymal stem cells for diabetes mellitus treatment: new advances. Biomedical Research and Therapy. 2017;4(1):1062-1081.
  62. Mizuno H, Tobita M, Uysal AC. Concise review: Adipose-derived stem cells as a novel tool for future regenerative medicine. Stem Cells. 2012 May;30:804-10.
  63. Chandra V, Swetha G, Muthyala S, Jaiswal AK, Bellare JR, Nair PD, et al. Islet-like cell aggregates generated from human adipose tissue derived stem cells ameliorate experimental diabetes in mice. PLoS One. 2011;6:e20615.
  64. Zang L, Hao H, Liu J, Li Y, Han W, Mu Y. Mesenchymal stem cell therapy in type 2 diabetes mellitus. Diabetol Metab Syndr. 2017 May 15;9:36.
  65. Inzucchi SE. Oral antihyperglycemic therapy for type 2 diabetes: scientific review. JAMA. 2002 Jan 16;287:360-72.
  66. Voltarelli JC, Couri CE, Oliveira MC, Moraes DA, Stracieri AB, Pieroni F, et al. Stem cell therapy for diabetes mellitus. Kidney Int Suppl (2011). 2011 Sep;1:94-8.
  67. Estrada EJ, Valacchi F, Nicora E, Brieva S, Esteve C, Echevarria L, et al. Combined treatment of intrapancreatic autologous bone marrow stem cells and hyperbaric oxygen in type 2 diabetes mellitus. Cell Transplant. 2008;17:1295-304.
  68. Bhansali A, Upreti V, Khandelwal N, Marwaha N, Gupta V, Sachdeva N, et al. Efficacy of autologous bone marrow-derived stem cell transplantation in patients with type 2 diabetes mellitus. Stem Cells Dev. 2009 Dec;18:1407-16.
  69. Si Y, Zhao Y, Hao H, Liu J, Guo Y, Mu Y, 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 Jun;61:1616-25.
  70. Jiang R, Han Z, Zhuo G, Qu X, Li X, Wang X, et al. Transplantation of placenta-derived mesenchymal stem cells in type 2 diabetes: a pilot study. Front Med. 2011 Mar;5:94-100.
  71. Skyler JS, Fonseca VA, Segal KR, Rosenstock J; MSB-DM003 Investigators. Allogeneic Mesenchymal Precursor Cells in Type 2 Diabetes: A Randomized, Placebo-Controlled, Dose-Escalation Safety and Tolerability Pilot Study. Diabetes Care. 2015 Sep;38:1742-9.
  72. Wu Z, Cai J, Chen J, Huang L, Wu W, Luo F, et al. Autologous bone marrow mononuclear cell infusion and hyperbaric oxygen therapy in type 2 diabetes mellitus: an open-label, randomized controlled clinical trial. Cytotherapy. 2014 Feb;16:258-65.
  73. Wang L, Zhao S, Mao H, Zhou L, Wang ZJ, Wang HX. Autologous bone marrow stem cell transplantation for the treatment of type 2 diabetes mellitus. Chin Med J (Engl). 2011 Nov;124:3622-8.
  74. Bhansali S, Dutta P, Kumar V, Yadav MK, Jain A, Mudaliar S, et al. Efficacy of Autologous Bone Marrow-Derived Mesenchymal Stem Cell and Mononuclear Cell Transplantation in Type 2 Diabetes Mellitus: A Randomized, Placebo-Controlled Comparative Study. Stem Cells Dev. 2017 Apr 1;26:471-81.
  75. Kempen JH, O'Colmain BJ, Leske MC, Haffner SM, Klein R, Moss SE, et al. Eye Diseases Prevalence Research Group. The prevalence of diabetic retinopathy among adults in the United States. Arch Ophthalmol. 2004 Apr;122:552-63.
  76. Frank RN. Diabetic retinopathy. N Engl J Med. 2004 Jan 1;350:48-58.
  77. Calcutt NA, Cooper ME, Kern TS, Schmidt AM. Therapies for hyperglycaemia-induced diabetic complications: from animal models to clinical trials. Nat Rev Drug Discov. 2009 May;8:417-29.
  78. Lee IG, Chae SL, Kim JC. Involvement of circulating endothelial progenitor cells and vasculogenic factors in the pathogenesis of diabetic retinopathy. Eye (Lond). 2006 May;20:546-52.
  79. Brunner S, Schernthaner GH, Satler M, Elhenicky M, Hoellerl F, Schmid-Kubista KE, et al. Correlation of different circulating endothelial progenitor cells to stages of diabetic retinopathy: first in vivo data. Invest Ophthalmol Vis Sci. 2009 Jan;50:392-8.
  80. Ciulla TA, Amador AG, Zinman B. Diabetic retinopathy and diabetic macular edema: pathophysiology, screening, and novel therapies. Diabetes Care. 2003 Sep;26:2653-64.
  81. Solmaz V, Tekatas A, Erdoğan MA, Erbaş O. Exenatide, a GLP-1 analog, has healing effects on LPS-induced autism model: Inflammation, oxidative stress, gliosis, cerebral GABA, and serotonin interactions. Int J Dev Neurosci. 2020 Nov;80:601-12.
  82. Inoue Y, Iriyama A, Ueno S, Takahashi H, Kondo M, Tamaki Y, et al. Subretinal transplantation of bone marrow mesenchymal stem cells delays retinal degeneration in the RCS rat model of retinal degeneration. Exp Eye Res. 2007 Aug;85:234-41.
  83. Wang S, Lu B, Girman S, Duan J, McFarland T, Zhang QS, et al. Non-invasive stem cell therapy in a rat model for retinal degeneration and vascular pathology. PLoS One. 2010 Feb 15;5:e9200.
  84. Scalinci SZ, Scorolli L, Corradetti G, Domanico D, Vingolo EM, Meduri A, et al. Potential role of intravitreal human placental stem cell implants in inhibiting progression of diabetic retinopathy in type 2 diabetes: neuroprotective growth factors in the vitreous. Clin Ophthalmol. 2011;5:691-6.
  85. Mottaghi S, Larijani B, Sharifi AM. Atorvastatin: an efficient step forward in mesenchymal stem cell therapy of diabetic retinopathy. Cytotherapy. 2013 Mar;15:263-6.
  86. Siqueira RC, Messias A, Gurgel VP, Simões BP, Scott IU, Jorge R. Improvement of ischaemic macular oedema after intravitreal injection of autologous bone marrow-derived haematopoietic stem cells. Acta Ophthalmol. 2015 Mar;93:e174-6.
  87. Grant MB, May WS, Caballero S, Brown GA, Guthrie SM, Mames RN, et al. Adult hematopoietic stem cells provide functional hemangioblast activity during retinal neovascularization. Nat Med. 2002 Jun;8:607-12.
  88. Jarajapu YP, Grant MB. The promise of cell-based therapies for diabetic complications: challenges and solutions. Circ Res. 2010 Mar 19;106:854-69.
  89. Ritter MR, Banin E, Moreno SK, Aguilar E, Dorrell MI, Friedlander M. Myeloid progenitors differentiate into microglia and promote vascular repair in a model of ischemic retinopathy. J Clin Invest. 2006 Dec;116:3266-76.
  90. Zhou JY, Zhang Z, Qian GS. Mesenchymal stem cells to treat diabetic neuropathy: a long and strenuous way from bench to the clinic. Cell Death Discov. 2016 Jul 11;2:16055.
  91. Han JW, Sin MY, Yoon YS. Cell therapy for diabetic neuropathy using adult stem or progenitor cells. Diabetes Metab J. 2013 Apr;37:91-105.
  92. Lupachyk S, Shevalye H, Maksimchyk Y, Drel VR, Obrosova IG. PARP inhibition alleviates diabetes-induced systemic oxidative stress and neural tissue 4-hydroxynonenal adduct accumulation: correlation with peripheral nerve function. Free Radic Biol Med. 2011 May 15;50:1400-9.
  93. Cameron NE, Cotter MA. Effects of antioxidants on nerve and vascular dysfunction in experimental diabetes. Diabetes Res Clin Pract. 1999 Sep;45:137-46.
  94. Hortu I, Ozceltik G, Sahin C, Akman L, Yildirim N, Erbas O. Granulocyte Colony-Stimulating Factor Prevents Ischemia/Reperfusion-Induced Ovarian Injury in Rats: Evaluation of Histological and Biochemical Parameters. Reprod Sci. 2019 Oct;26:1389-94.
  95. Shibata T, Naruse K, Kamiya H, Kozakae M, Kondo M, Yasuda Y, et al. Transplantation of bone marrow-derived mesenchymal stem cells improves diabetic polyneuropathy in rats. Diabetes. 2008 Nov;57:3099-107.
  96. Hao H, Liu J, Shen J, Zhao Y, Liu H, Hou Q, et al. Multiple intravenous infusions of bone marrow mesenchymal stem cells reverse hyperglycemia in experimental type 2 diabetes rats. Biochem Biophys Res Commun. 2013 Jul 5;436:418-23.
  97. Cavusoglu T, Karadeniz T, Cagiltay E, Karadeniz M, Yigitturk G, Acikgoz E, et al. The protective effect of losartan on diabetic neuropathy in a diabetic rat model. Exp Clin Endocrinol Diabetes. 2015 Sep;123(8):479-84.
  98. Sakar M, Korkusuz P, Demirbilek M, Cetinkaya DU, Arslan S, Denkbaş EB, et al. The effect of poly(3-hydroxybutyrate-co-3- hydroxyhexanoate) (PHBHHx) and human mesenchymal stem cell (hMSC) on axonal regeneration in experimental sciatic nerve damage. Int J Neurosci. 2014 Sep;124:685-96.
  99. Dronavalli S, Duka I, Bakris GL. The pathogenesis of diabetic nephropathy. Nat Clin Pract Endocrinol Metab. 2008 Aug;4:444-52.
  100. Yamagishi S, Matsui T. Advanced glycation end products, oxidative stress and diabetic nephropathy. Oxid Med Cell Longev. 2010 Mar-Apr;3:101-8.
  101. Kanwar YS, Sun L, Xie P, Liu FY, Chen S. A glimpse of various pathogenetic mechanisms of diabetic nephropathy. Annu Rev Pathol. 2011;6:395-423.
  102. Mathieson PW. The podocyte as a target for therapies--new and old. Nat Rev Nephrol. 2011 Nov 1;8:52-6.
  103. Gilbert RE, Cooper ME. The tubulointerstitium in progressive diabetic kidney disease: more than an aftermath of glomerular injury? Kidney Int. 1999 Nov;56:1627-37.
  104. Grgic I, Campanholle G, Bijol V, Wang C, Sabbisetti VS, Ichimura T, et al. Targeted proximal tubule injury triggers interstitial fibrosis and glomerulosclerosis. Kidney Int. 2012 Jul;82:172-83.
  105. Fukami K, Yamagishi S, Ueda S, Okuda S. Role of AGEs in diabetic nephropathy. Curr Pharm Des. 2008;14:946-52.
  106. Bi B, Schmitt R, Israilova M, Nishio H, Cantley LG. Stromal cells protect against acute tubular injury via an endocrine effect. J Am Soc Nephrol. 2007 Sep;18:2486-96.
  107. Pala HG, Pala EE, Artunc Ulkumen B, Aktug H, Yavasoglu A, Korkmaz HA, et al. The protective effect of granulocyte colony-stimulating factor on endometrium and ovary in a rat model of diabetes mellitus. Gynecol Obstet Invest. 2014;78:94-100.
  108. Song B, Smink AM, Jones CV, Callaghan JM, Firth SD, Bernard CA, et al. The directed differentiation of human iPS cells into kidney podocytes. PLoS One. 2012;7:e46453.
  109. Lam AQ, Freedman BS, Morizane R, Lerou PH, Valerius MT, Bonventre JV. Rapid and efficient differentiation of human pluripotent stem cells into intermediate mesoderm that forms tubules expressing kidney proximal tubular markers. J Am Soc Nephrol. 2014 Jun;25:1211-25.
  110. Nagaishi K, Mizue Y, Chikenji T, Otani M, Nakano M, Konari N, et al. Mesenchymal stem cell therapy ameliorates diabetic nephropathy via the paracrine effect of renal trophic factors including exosomes. Sci Rep. 2016 Oct 10;6:34842.
  111. Ezquer FE, Ezquer ME, Parrau DB, Carpio D, Yañez AJ, Conget PA. Systemic administration of multipotent mesenchymal stromal cells reverts hyperglycemia and prevents nephropathy in type 1 diabetic mice. Biol Blood Marrow Transplant. 2008 Jun;14:631-40.
  112. Ezquer F, Giraud-Billoud M, Carpio D, Cabezas F, Conget P, Ezquer M. Proregenerative Microenvironment Triggered by Donor Mesenchymal Stem Cells Preserves Renal Function and Structure in Mice with Severe Diabetes Mellitus. Biomed Res Int. 2015;2015:164703.
  113. Bozkurt MF, Bhaya MN, Dibekoğlu C, Akat A, Ateş U, Erbaş O. Mesenchymal stem cells have ameliorative effect on the colitis model via Nrf2/HO-1 pathway. Acta Cir Bras. 2022 Oct 10;37:e370704.
  114. Bernardi S, Severini GM, Zauli G, Secchiero P. Cell-based therapies for diabetic complications. Exp Diabetes Res. 2012;2012:872504.
  115. Beckman JA, Creager MA, Libby P. Diabetes and atherosclerosis: epidemiology, pathophysiology, and management. JAMA. 2002 May 15;287:2570-81.
  116. Fadini GP, Miorin M, Facco M, Bonamico S, Baesso I, Grego F, et al. Circulating endothelial progenitor cells are reduced in peripheral vascular complications of type 2 diabetes mellitus. J Am Coll Cardiol. 2005 May 3;45:1449-57.
  117. Fadini GP, Sartore S, Albiero M, Baesso I, Murphy E, Menegolo M, et al. Number and function of endothelial progenitor cells as a marker of severity for diabetic vasculopathy. Arterioscler Thromb Vasc Biol. 2006 Sep;26:2140-6.
  118. Schmidt-Lucke C, Rössig L, Fichtlscherer S, Vasa M, Britten M, Kämper U, et al. Reduced number of circulating endothelial progenitor cells predicts future cardiovascular events: proof of concept for the clinical importance of endogenous vascular repair. Circulation. 2005 Jun 7;111:2981-7.
  119. Erbaş O, Altuntaş İ, Çağlar Ö, Özyilmaz E, Sari E, Üzümcü İ, et al. Experimental Model of Cardiotoxicity [Internet]. Risk Factors for Cardiovascular Disease. IntechOpen; 2022. Available from: http://dx.doi.org/10.5772/ intechopen.101401
  120. Khan ZA, Boscolo E, Picard A, Psutka S, Melero-Martin JM, Bartch TC, et al. Multipotential stem cells recapitulate human infantile hemangioma in immunodeficient mice. J Clin Invest. 2008 Jul;118:2592-9.
  121. Melero-Martin JM, Khan ZA, Picard A, Wu X, Paruchuri S, Bischoff J. In vivo vasculogenic potential of human blood-derived endothelial progenitor cells. Blood. 2007 Jun 1;109:4761-8.
  122. Zhang N, Li J, Luo R, Jiang J, Wang JA. Bone marrow mesenchymal stem cells induce angiogenesis and attenuate the remodeling of diabetic cardiomyopathy. Exp Clin Endocrinol Diabetes. 2008 Feb;116:104-11.
  123. Naruse K, Hamada Y, Nakashima E, Kato K, Mizubayashi R, Kamiya H, et al. Therapeutic neovascularization using cord blood-derived endothelial progenitor cells for diabetic neuropathy. Diabetes. 2005 Jun;54:1823-8.
  124. Huang P, Li S, Han M, Xiao Z, Yang R, Han ZC. Autologous transplantation of granulocyte colony-stimulating factor-mobilized peripheral blood mononuclear cells improves critical limb ischemia in diabetes. Diabetes Care. 2005 Sep;28:2155-60.