Stem Cells a better way for Treatment: Diabetes

Authors

  • Mamta Kumari Dayanand Dinanath College, Institute of Pharmacy, Kanpur Nagar-209214
  • Parmanand Pandey Nector Life Sciences, Jharmajari, Baddi-173205 Himanchal Pradesh
  • Anupam Kr Sachan Nector Life Sciences, Jharmajari, Baddi-173205 Himanchal Pradesh, India

DOI:

https://doi.org/10.22270/ajprd.v9i1.911

Keywords:

Stem cell, Cell therapy, Diabetes, Insulin

Abstract

Insulin-producing cells derived from the stem cell embryonic stem cell  and pluripotent stem cell have been long duration to encourage, but evasive treatment far from clinical interpret into type1 diabetes therapy.  Although stem cell therapies provide a great opportune time there is also conceivable risk such as teratoma formation to relate with the treatment. Mesenchyme stem stromal cells have due to their modulator effects on immunity, inflammation, and tissue repair been suggested to be used to either halt beta-cell loss during T1D development or be used to protect and support pancreatic islets when transplanted. This review aims to give an overview of the current knowledge of stem cell therapy outcomes in animal models of type-1diabetes and a proposed road map towards the clinical setting with a special focus on the potential risks and hurdles which need to be considered. From a clinical point of view, transplantation of insulin-producing cells derived from stem cells must be performed without immune suppression to be an attractive treatment option.

 

Downloads

Download data is not yet available.

Author Biographies

Mamta Kumari, Dayanand Dinanath College, Institute of Pharmacy, Kanpur Nagar-209214

Dayanand Dinanath College, Institute of Pharmacy, Kanpur Nagar-209214

Parmanand Pandey, Nector Life Sciences, Jharmajari, Baddi-173205 Himanchal Pradesh

Nector Life Sciences, Jharmajari, Baddi-173205 Himanchal Pradesh

Anupam Kr Sachan, Nector Life Sciences, Jharmajari, Baddi-173205 Himanchal Pradesh, India

Nector Life Sciences, Jharmajari, Baddi-173205 Himanchal Pradesh, India

References

1. Sachan AK, Ch. V. Rao, Sachan NK, Determination of Antidiabetic Potential in Crude Extract of Caesalpinia bonducella Wild on normal and Streptozotocin Induced Diabetic Rats. Research J. Pharm. and Tech 2020.00162.6
2. Sachan AK, Rao ChV, Sachan NK, An Investigation into phytochemical composition of Caesalpinia bonducella Wild from Chambal Valley, India. World Appl Sci J 2017; 35 6: 896-901.
3. Sachan AK, Rao ChV, Sachan NK, Ethnobotanical survey of indigenous medicines practiced in Chambal Valley of Uttar Pradesh. NISCAIR-CSIR: Bharatiya Vaigyanik evam Audyogik Anusandhan Patrika 2015; 232: 132-135.
4. Sheweita SA, Mashaly S, Newairy AA, et al. Changes in oxidative stress and antioxidant enzyme activities in streptozotocin-induced diabetes mellitus in rats: Role of Alhagi maurorum Extracts. Oxidative Medicine and Cellular Longevity, Hindawi Publishing Corporation 2016; 1-8.
5. Wild S, Roglic G, Green A, et al. Global prevalence of diabetes: Estimates for the year 2000 and projections for 2030. Diabetes Care 2004; 27:1047-53.
6. Secrete R, Shaw J, Zimmet P, et al. Diabetes, and impaired glucose tolerance. In: Gan D, editor. Diabetes Atlas 2016; 1-8.
7. International Diabetes Federation, et al. 3rd ed. Belgium: International Diabetes Federation; 2006.
8. Caplan AI, DA Ede, JR Hinchliffe, et al. Muscle, cartilage, and bone development and differentiation from chick limb mesenchymal cells in-Vertebrate Limb and Somite Morphogenesis, ed by, Cambridge, England, Cambridge University Press, 1977-199-213.
9. Zipori .D and Lee F, Introduction of interleukin-3 gene into stroma1 cells from the bone marrow alters hematopoietic differentiation but does not modify stem cell renewal. Blood 1988-71586596.
10. Baron R , Caplan M ,Neff L , et al. The molecular control of muscle and cartilage development. In: 39th Annual Symposium of the Society for Developmental Biology, ed by S Subtelney, U Abbott, New York, Alan R. Liss, 1981-37-68.
11. Weissman IL, Stem cells: units of development, units of regeneration, and units in evolution. Cell. 2000; 100:157–68.
12. Pittenger MF, A M Mackay, S C Beck, et al. Multilineage potential of adult human mesenchymal stem cells. Science. 1999; 284:143–7.
13. Nikulina, S.E., Ciaraldi, T.P., Mudaliar, S., et al. Potential role of glycogen synthase kinase-3 in skeletal muscle insulin resistance of type 2 diabetes. Diabetes 2000-49, 263271.
14. Sethi, J.K., Vidal-Puig, A.,et al. Wnt signaling and the control of cellular metabolism. Biochem. J. 2010-427, 1e17.
15. Janich, P and Pascual, G, The circadian molecular clock creates epidermal stem cell heterogeneity. Nature 2009- 2011. 480-214.
16. L.T. Lock and E.S, Tzanakakis, Stem/progenitor cell sources of insulin-producing cells for the treatment of diabetes, Tissue Eng. 13 2007 1399–1412.
17. C.J. Schoenherr and D.J. Anderson, The neuron-restrictive silencer factor NRSF: a coordinate repressor of multiple neuron-specific genes, Science 267 1995 1360–1363.
18. Blyszczuk, P and Czyz, J, Expression of Pax4 in embryonic stem cells promotes differentiation of nestin-positive progenitor and insulin-producing cells. Proc. Natl. Acad. Sci. U.S.A. 2003-100, 998–1003 86.
19. Satsuki Miyazaki , Eiji Yamato, Jun-ichi Miyazaki , Regulated expression of pdf-1 promotes in vitro differentiation of insulin-producing cells from embryonic stem cells. Diabetes 2004 53,-1030–1037.
20. Leon-Quinto, T., Jones, J., et al. In vitro directed differentiation of mouse embryonic stem cells into insulin-producing cells. Diabetologia 2004 -47, 1442–1451.
21. H. Baharvand, and H. Jafary, Generation of insulin-secreting cells from human undeveloped embryonic stem cells, Dev. Development. Contrast 2006-48 323e332.
22. G.H. Mao, G.A. Chen, H.Y. Bai, et al. The inversion of hyperglycemia in diabetic mice utilizing PLGA frameworks cultivated with islet-like cells got from human embryonic stem cell, Biomaterials 30 2009-1706e1714.
23. Teo AK, Wagers AJ, Kulkarni RN, et al. New opportunities: harnessing induced pluripotency for discovery in diabetes and metabolism. Cell Metab. 2013-18:775–91.
24. Russ HA, Parent AV, Ringler JJ, et al. Controlled enlistment of human pancreatic ancestors produces useful beta-like cells in vitro. EMBO J. 2015; 34:1759–72.
25. Clark GO, Yochem RL, Axelman J, et al. Glucose responsive insulin creation from human early-stage germ EG cell subsidiaries. Biochim Biophys Res Commun. 2007;356:587–9.
26. Soleimanpour SA, Ferrari AM, Raum JC, et al. Diabetes susceptibility genes Pdx1 and Clec16a function in a pathway regulating mitophagy in β-cells. Diabetes. 2015;64:3475–3484.
27. McKenna B, Guo M, Reynolds A, et al. Dynamic recruitment of functionally distinct Swi/Snf chromatin remodeling complexes modulate Pdx1 activity in islet β cells. Cell Rep. 2015;10:2032–2042.
28. Wang X, Lei XG, Wang J, et al. Malondialdehyde regulates glucose-stimulated insulin secretion in murine islets via the TCF7L2-dependent Wnt signaling pathway. Mol Cell Endocrinol. 2014;382:8–16.
29. Burrows GG, Vanʼt Hof W, Newell LF, et al. Dissection of the human multipotent adult progenitor cell secretome by proteomic analysis. Stem Cells Transl Med 2013 2:745–757.
30. Daoud J, Rosenberg L, Tabrizian M, et al. Pancreatic islet culture and preservation strategies: advances, challenges, and future outlook. Cell Transplant 2010 19:1523.
31. Stubbs SL, Hsiao ST, Peshavariya HM, et al. Hypoxic preconditioning enhances the survival of human adipose-derived stem cells and conditions endothelial cells in vitro. Stem cells and development. 2012; 2111:1887–96.
32. Krampera M, Cosmi L, Angeli R, et al. Role for interferon-gamma in the immunomodulatory activity of human bone marrow mesenchymal stem cells. Stem cells. 2006; 24 2:386–98.
33. Yao Y, Zhang F, Wang L, et al. Lipopolysaccharide preconditioning enhances the efficacy of mesenchymal stem cell transplantation in a rat model of acute myocardial infarction. Journal of biomedical science. 2009; 16:74
34. Arden, G. B. and Sivaprasad.S, The pathogenesis of early retinal changes of diabetic retinopathy. Doc Ophthalmol 124, 15–26 2012.
35. Si, Y. L., Zhao, Y. L.Hao, H. J., et al. MSCs: biological characteristics, clinical applications, and their outstanding concerns. Ageing Res Rev 10, 93–103 2011.
36. Zhang, W., Wang, Y., Kong, J. et al. Roles of Wnt/β-catenin signaling in retinal neuron-like differentiation of bone marrow mesenchymal stem cells from nonobese diabetic mice. J Mol Neurosci 49, 250–61 2013.
37. Zhang R, and Lipasin, a novel nutritionally-regulated liver-enriched factor that regulates serum triglyceride levels. Biochim Biophys Res Commun. 424:786–792. 2012.

38. Quagliarini F, Wang Y, Kozlitina J, et al. Atypical angiopoietin-like protein that regulates ANGPTL3. Proc Natl AcadSci USA. 109:19751–19756. 2012.
39. Li X.and MiR-375, et al. A microRNA related to diabetes. Gene 2014; 533:1e4.
40. Poy MN, Eliasson L, Krutz,feldt J, et al. A pancreatic islet-specific microRNA regulates insulin secretion. Nature 2004;432:226e30
41. Dai R, Wang Z, Samanipour R, et al. Adipose-derived stem cells for tissue engineering and regenerative medicine applications. Stem Cells Int 2016;2016 :6737345.
42. Wankhade UD, Shen M, Kolhe R,et al. Advances in adipose-derived stem cell isolation, characterization, and application in regenerative tissue engineering. Stem Cells Int 2016; 2016:3206807.
43. Trounson A, DeWitt ND, Feigal EG, et al. The Alpha Stem Cell Clinic: A model for evaluating and delivering stem cell-based therapies. Stem cells translational medicine 2012; 1:9–14.
44. Zuk PA, Zhu M, Mizuno H, et al. Multilineage cells from human fat tissue: Implications for cell-based treatments. Tissue Eng 2001;7:211–228.
45. Konermann S, Brigham MD, Trevino AE, et al. Genome-scale transcriptional actuation by a designed CRISPR-Cas9 complex. Nature. 2015;5177536:583.8 .
46. Gimenez CA, Ielpi M, Mutto A, et al. CRISPR-on framework for the enactment of the endogenous human INS quality. Quality There. 2016;236:543.7.
47. Black Joshua B, Adler Andrew F, Wang, et al. Directed epigenetic renovating of endogenous loci by CRISPR/Cas9-based transcriptional activators straightforwardly changes fibroblasts over to neuronal cells. Cell Stem Cell. 2016; 193:406. 14. 49.
48. Kusiak B, Cleto S, Perez-Pinera P, et al. Engineering synthetic gene circuits in living cells with CRISPR technology. Trends Biotechnology. 2016; 347:535 .47.
49. Li X, Ling.Q, Wang Y, et al. Human Placenta-Derived Adherent Cells Prevent Bone Loss, Stimulate Bone Formation, and Suppress Growth of Multiple Myeloma in Bone. Stem Cells 2011; 29:263-73.
50. Abdi R, Fiorina P, Adra CN, et al. Immunomodulation by mesenchyme stem cells: a potential therapeutic strategy for type 1 diabetes. Diabetes 2008; 57:1759-67.
51. Lechner A, Yang YG, Blacken RA, et al. No evidence for significant transdifferentiation of bone marrow into pancreatic ß-cells in vivo. Diabetes 2004;53:616-23.
52. Figliuzzi M, Bonandrini B, Silvani S, et al. Mesenchymal stem cells help pancreatic islet transplantation to control type 1 diabetes. World J Stem Cells 2014; 6:16372.
53. Hematite P, Jaehyup K, Stein AP, et al. Potential role of mesenchymal stromal cells in pancreatic islet transplantation. Transplant Rev 2013; 27:21-9.
54. Gruessner AC and Sutherland DE. Pancreas transplant outcomes for United States US and non-US cases as reported to the United Network for Organ Sharing UNOS and the International Pancreas Transplant Registry IPTR as of June Clin Transplant. 2005; 19:433–455.
55. Alejandro R, Barton FB, Hering BJ,et al. Collaborative Islet Transplant Registry 2008 Update from the Collaborative Islet Transplant Registry. Transplantation. 2008; 86:17831788.
56. Nathan DM, Zinman B, Cleary PA, Orchard TJ 2009; 169:1307–1316.
57. Abdi R, Fiorina P, Adra CN, et al. Immunomodulation by mesenchymal stem cells: a potential therapeutic strategy for type 1 diabetes. Diabetes. 2008; 57:1759–1767.
58. Hess D, Li L, Martin M, et al. Bone marrow-derived stem cells initiate pancreatic regeneration. Nat Biotechnol. 2003;21:763–770.
59. 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. ProcNatlAcadSci USA. 2006;103:17438–17443.
60. Tseng YH, Cypress AM, Kahn CR, et al. Cellular bioenergetics as a target for obesity therapy. Nat Rev Drug Discover. 2010;9:465–482.
61. Scharp DW, Lacy PE, Santiago JV, et al. Results from our first nine intraportal islet allografts in type 1, insulin-dependent diabetic patients. Transplantation. 1991;51:76–85.
62. Sachs DH and Bonner-Weir S 2000, New islets from old. Nat Med 6: 250-251.
63. 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.
64. Rackham CL, Chagastelles PC, Nardi NB, Co-transplantation of mesenchymal stem cells maintain islet organization and morphology in mice. Diabetologia. 2011;54:1127–1135.
65. Gabr MM, Zakaria MM, Refaie AF, et al. Insulin-producing cells from adult human bone marrow mesenchymal stem cells control streptozotocin-induced diabetes in nude mice. Cell Transplant2012;21:997–1009.
66. Caplan AI and Dennis JE, Mesenchymal stem cells are trophic mediators. J Cell Brioche. 2006; 985:1076–84.
67. Chen L, Tredget EE, Wu PY, et al. Peregrine factors in mesenchymal stem cells recruit’s macrophages and endothelial lineage cells to enhance wound healing. PLoS ONE. 2008; 34:e1886.
68. Yi P, and Melton DA, Betatrophin: A hormone that controls pancreatic β cell proliferation. Cell. 153:747–758. 2013.

Published

2021-02-15

How to Cite

Kumari, M., Pandey, P., & Sachan, A. K. (2021). Stem Cells a better way for Treatment: Diabetes. Asian Journal of Pharmaceutical Research and Development, 9(1), 190–197. https://doi.org/10.22270/ajprd.v9i1.911