Int J Stem Cells.  2017 Nov;10(2):129-143. 10.15283/ijsc17035.

Combination of Obestatin and Bone Marrow Mesenchymal Stem Cells Prevents Aggravation of Endocrine Pancreatic Damage in Type II Diabetic Rats

Affiliations
  • 1Department of Physiology, Faculty of Medicine, Benha University, Cairo, Egypt.
  • 2Department of Histology, Faculty of Medicine, Benha University, Cairo, Egypt.
  • 3Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Cairo, Egypt. dinasabry@kasralainy.edu.eg

Abstract

One of the new promising therapies in treatment of diabetes mellitus is mesenchymal stem cells (MSCs) which have an interesting therapeutic potentiality based on their paracrine effect and transdifferentiation potentiality. Also obestatin improves the generation of functional β cells/islet-like cell clusters in vitro, suggesting implications for cell-based replacement therapy in diabetes. So the aim of this study was to evaluate the effect of combination of both MSCs and obestatin on an experimental model of type II diabetes mellitus (T2DM). Sixty male rats were divided into; group I (control group), group II (T2DM group) induced by administration of high fat diet (HFD) and injection of streptozotocin (STZ) in low dose, group III (T2DM treated with MSCs), group IV (T2DM treated with obestatin), group V (T2DM treated with MSCs and obestatin). Fasting blood glucose, C-peptide, insulin and lipid profile were measured. HOMA-IR and HOMA-β were calculated. Pancreatic expression of insulin, glucagon like peptide -1 (GLP-1) and pancreatic duodenal homeobox 1 (Pdx1) mRNA levels were measured. In addition pancreatic histological changes, insulin and Bax were analyzed by immunohistochemical examination of islets of Langerhans. Diabetic rats showed significant increase in HOMA-IR, serum glucose and lipid profile levels with significant decrease in insulin, HOMA-β, GLP-1 and Pdx1 levels. MSCs and obestatin caused significant improvement in all parameters with more significant improvement in combined therapy. The protective effects afforded by MSCs and obestatin may derive from improvement of the metabolic profile, antiapoptosis and by increase in pancreatic GLP-1and Pdx1 gene expression.

Keyword

Mesenchymal stem cells; Obestatin; Type II diabetes mellitus

MeSH Terms

Animals
Blood Glucose
Bone Marrow*
C-Peptide
Diabetes Mellitus
Diet, High-Fat
Fasting
Gene Expression
Genes, Homeobox
Ghrelin*
Glucagon
Glucagon-Like Peptide 1
Humans
In Vitro Techniques
Insulin
Islets of Langerhans
Male
Mesenchymal Stromal Cells*
Metabolome
Models, Theoretical
Rats*
RNA, Messenger
Streptozocin
Blood Glucose
C-Peptide
Ghrelin
Glucagon
Glucagon-Like Peptide 1
Insulin
RNA, Messenger
Streptozocin

Figure

  • Fig. 1 Homing of MSCs in pancreas. (A) Primary culture of MSCs showing many spindle-shaped stem cells (white arrows) ×200. (B) Fluorescent microscopic image of a section in pancreas of rat in group III demonstrating the green fluorescence of MSCs labeled with GFP two week after implantation (white arrows) ×200. (C) Flow cytometric chart analysis for surface antigens of MSCs. They were positive for CD29 and CD90.

  • Fig. 2 Photomicrographs sections (immunohistochemistry) from rat tail pancreas showing. (A) GFP expression in transplanted MSCs. (B) CD105 expression in transplanted MSCs.

  • Fig. 3 Photomicrographs sections (H&E) from rat tail pancreas showing. (A) A section from control rat illustrating pancreatic lobules (bent arrows) separated by thin connective tissue septae (black arrow heads), islet of Langerhans (white arrow head), blood vessels (white ↑) and pancreatic ducts (black ↑)×100. (B) A section from a control rat revealed an islet of Langerhans (black arrow head) containing β cells in the center (white arrow head) and most probably α cells in the periphery (bent arrow), blood vessel (black ↑) and pancreatic acini formed of pyramidal cells with basal basophilia and apical acidophilia (white↑)×400. (C) A section of a rat from group II revealed two shrunken distorted islets of Langerhans with marked loss of their cells (arrow head), some cells with vacuolated cytoplasm (↑) and darkly stained nuclei. Note distorted pancreatic acini (bent arrow)×400. (D) A section of a rat from group III showing an islet of Langerhans (arrow head) with vacuolated cells (↑) surrounded by exocrine pancreatic acini ×400. (E) A section of a rat from group IV showing an islet of Langerhans (arrow head) and some cells with vacuolated cytoplasm (bent arrows)×400. (F) A section of a rat from group V revealed well-defined islet of Langerhans (arrow head) composed of cords of endocrine cells which are small with a pale-stained granular cytoplasm, exocrine pancreatic acini (bent arrows) and small blood capillaries (↑)×400.

  • Fig. 4 Photomicrographs sections in the tail of pancreas revealed islets of Langerhans (immunohistochemistry anti-insulin antibodies ×400) showing. (A) Strong positively stained secretory granules of the β cells in a control rat. (B) Mild positively stained secretory granules of the β cells in a rat from T2DM group. (C) Moderate positively stained secretory granules of β cells of a rat from T2DM+MSCs group. (D) Moderate positively stained secretory granules of β cells in a rat from T2DM+obestatin group. (E) A highly positive stained secretory granules of β cells in a rat from T2DM+MSCs+obestatin group.

  • Fig. 5 Photomicrographs of sections in the tail of pancreas revealed islets of Langerhans (immunohistochemistry anti-Bax antibodies ×400) showing. (A) Negative immunostaining of the β cells in a control rat. (B) Highly positive Bax immunostaining in the islet of Langerhans indicating increasing rate of cellular apoptosis in a rat from T2DM group. (C) Weak positive Bax immunostaining in the islet of Langerhans in a rat from T2DM+MSCs group. (D) Weak positive Bax immunostaining in the islet of Langerhans in a rat from T2DM+obestatin group. (E) Minimally expressed Bax immunostaining in the islet of Langerhans in a rat from T2DM+MSCs+obestatin group.

  • Fig. 6 The mean area percentages of insulin and Bax immunostaining for all groups. aSignificant difference (p<0.05) compared with control group. bSignificant difference (p<0.05) compared with T2DM group. cSignificant difference (p<0.05) compared with T2DM+MSCs group. dSignificant difference (p<0.05) compared with T2DM+obestatin group. eSignificant difference (p<0.05) compared with T2DM+MSCs+obestatin group.


Reference

References

1. Ma RCW, Tong PCY. Epidemiology of type 2 diabetes. Holt RIG, Cockram CS, Flyvbjerg A, Goldstein BJ, editors. Textbook of Diabetes. 5th ed. ch 4. USA: JohnWiley & Sons Ltd;2017. p. 43–64.
2. Stumvoll M, Goldstein BJ, van Haeften TW. Type 2 diabetes: principles of pathogenesis and therapy. Lancet. 2005; 365:1333–1346. DOI: 10.1016/S0140-6736(05)61032-X. PMID: 15823385.
Article
3. Asghar Z, Yau D, Chan F, Leroith D, Chan CB, Wheeler MB. Insulin resistance causes increased beta-cell mass but defective glucose-stimulated insulin secretion in a murine model of type 2 diabetes. Diabetologia. 2006; 49:90–99. DOI: 10.1007/s00125-005-0045-y.
Article
4. Fujimoto K, Polonsky KS. Pdx1 and other factors that regulate pancreatic beta-cell survival. Diabetes Obes Metab. 2009; 11(Suppl 4):30–37. DOI: 10.1111/j.1463-1326.2009.01121.x. PMID: 19817786. PMCID: 2802270.
5. Zenari L, Marangoni A. What are the preferred strategies for control of glycaemic variability in patients with type 2 diabetes mellitus? Diabetes Obes Metab. 2013; 15(Suppl 2):17–25. DOI: 10.1111/dom.12143. PMID: 24034516.
Article
6. Ren AJ, Guo ZF, Wang YK, Lin L, Zheng X, Yuan WJ. Obestatin, obesity and diabetes. Peptides. 2009; 30:439–444. DOI: 10.1016/j.peptides.2008.10.002.
Article
7. Gesmundo I, Gallo D, Favaro E, Ghigo E, Granata R. Obestatin: a new metabolic player in the pancreas and white adipose tissue. IUBMB Life. 2013; 65:976–982. DOI: 10.1002/iub.1226. PMID: 24217898.
Article
8. Granata R, Gallo D, Luque RM, Baragli A, Scarlatti F, Grande C, Gesmundo I, Córdoba-Chacón J, Bergandi L, Settanni F, Togliatto G, Volante M, Garetto S, Annunziata M, Chanclón B, Gargantini E, Rocchietto S, Matera L, Datta G, Morino M, Brizzi MF, Ong H, Camussi G, Castaño JP, Papotti M, Ghigo E. Obestatin regulates adipocyte function and protects against diet-induced insulin resistance and inflammation. FASEB J. 2012; 26:3393–3411. DOI: 10.1096/fj.11-201343. PMID: 22601779.
Article
9. Baragli A, Grande C, Gesmundo I, Settanni F, Taliano M, Gallo D, Gargantini E, Ghigo E, Granata R. Obestatin enhances in vitro generation of pancreatic islets through regulation of developmental pathways. PLoS One. 2013; 8:e64374. DOI: 10.1371/journal.pone.0064374. PMID: 23741322. PMCID: 3669302.
Article
10. Lévesque JP, Winkler IG, Larsen SR, Rasko JE. Mobilization of bone marrow-derived progenitors. Handb Exp Pharmacol. 2007; (180):3–36. DOI: 10.1007/978-3-540-68976-8_1. PMID: 17554502.
Article
11. Yeung TY, Seeberger KL, Kin T, Adesida A, Jomha N, Shapiro AM, Korbutt GS. Human mesenchymal stem cells protect human islets from pro-inflammatory cytokines. PLoS One. 2012; 7:e38189. DOI: 10.1371/journal.pone.0038189. PMID: 22666480. PMCID: 3364233.
Article
12. Boumaza I, Srinivasan S, Witt WT, Feghali-Bostwick C, Dai Y, Garcia-Ocana A, Feili-Hariri M. Autologous bone marrow-derived rat mesenchymal stem cells promote PDX-1 and insulin expression in the islets, alter T cell cytokine pattern and preserve regulatory T cells in the periphery and induce sustained normoglycemia. J Autoimmun. 2009; 32:33–42. DOI: 10.1016/j.jaut.2008.10.004.
Article
13. Si Y, Zhao Y, Hao H, Liu J, Guo Y, Mu Y, Shen J, Cheng Y, Fu X, Han W. 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: 10.2337/db11-1141. PMID: 22618776. PMCID: 3357293.
Article
14. Granata R, Volante M, Settanni F, Gauna C, Ghé C, Annunziata M, Deidda B, Gesmundo I, Abribat T, van der Lely AJ, Muccioli G, Ghigo E, Papotti M. Unacylated ghrelin and obestatin increase islet cell mass and prevent diabetes in streptozotocin-treated newborn rats. J Mol Endocrinol. 2010; 45:9–17. DOI: 10.1677/JME-09-0141. PMID: 20382773.
Article
15. Srinivasan K, Patole PS, Kaul CL, Ramarao P. Reversal of glucose intolerance by by pioglitazone in high fat diet-fed rats. Methods Find Exp Clin Pharmacol. 2004; 26:327–333. DOI: 10.1358/mf.2004.26.5.831322. PMID: 15319810.
Article
16. Mansor LS, Gonzalez ER, Cole MA, Tyler DJ, Beeson JH, Clarke K, Carr CA, Heather LC. Cardiac metabolism in a new rat model of type 2 diabetes using high-fat diet with low dose streptozotocin. Cardiovasc Diabetol. 2013; 12:136–146. DOI: 10.1186/1475-2840-12-136. PMID: 24063408. PMCID: 3849358.
Article
17. Hoybergs YM, Meert TF. The effect of low-dose insulin on mechanical sensitivity and allodynia in type I diabetes neuropathy. Neurosci Lett. 2007; 417:149–154. DOI: 10.1016/j.neulet.2007.02.087. PMID: 17412508.
Article
18. Abdel aziz MT, El Asmar MF, Atta HM, Mahfouz S, Fouad HH, Roshdy NK, Rashed LA, Sabry D, Hassouna AA, Taha FM. Efficacy of mesenchymal stem cells in suppression of hepatocarcinorigenesis in rats: possible role of Wnt signaling. J Exp Clin Cancer Res. 2011; 30:49–60. DOI: 10.1186/1756-9966-30-49. PMID: 21545718. PMCID: 3113743.
Article
19. Yamazoe K, Mishima H, Torigoe K, Iijima H, Watanabe K, Sakai H, Kudo T. Effects of atelocollagen gel containing bone marrow-derived stromal cells on repair of osteochondral defect in a dog. J Vet Med Sci. 2007; 69:835–839. DOI: 10.1292/jvms.69.835. PMID: 17827891.
Article
20. Niki H, Hosokawa S, Nagaike K, Tagawa T. A new immunofluorostaining method using red fluorescence of PerCP on formalin-fixed paraffin-embedded tissues. J Immunol Methods. 2004; 293:143–151. DOI: 10.1016/j.jim.2004.07.009. PMID: 15541284.
Article
21. Novelli EL, Diniz YS, Galhardi CM, Ebaid GM, Rodrigues HG, Mani F, Fernandes AA, Cicogna AC, Novelli Filho JL. Anthropometrical parameters and markers of obesity in rats. Lab Anim. 2007; 41:111–119. DOI: 10.1258/002367707779399518. PMID: 17234057.
Article
22. Friedewald WT, Levy RI, Fredrickson DS. Estimation of the concentration of low-density lipoprotein cholesterol in plasma, without use of the preparative ultracentrifuge. Clin Chem. 1972; 18:499–502. PMID: 4337382.
Article
23. Cacho J, Sevillano J, de Castro J, Herrera E, Ramos MP. Validation of simple indexes to assess insulin sensitivity during pregnancy in Wistar and Sprague-Dawley rats. Am J Physiol Endocrinol Metab. 2008; 295:E1269–E1276. DOI: 10.1152/ajpendo.90207.2008. PMID: 18796548.
Article
24. Sun G, Bishop J, Khalili S, Vasdev S, Gill V, Pace D, Fitzpatrick D, Randell E, Xie YG, Zhang H. Serum visfatin concentrations are positively correlated with serum tri-acylglycerols and down-regulated by overfeeding in healthy young men. Am J Clin Nutr. 2007; 85:399–404. PMID: 17284735.
Article
25. Bancroft JD, Layton C. The hematoxylin and eosin, connective and mesenchymal tissues with their stains. Suvarna SK, Layton C, Bancroft JD, editors. Bancroft’s Theory and Practice of Histological Techniques. 7th ed. Philadelphia: Churchill Livingstone;2013. p. 173–212. DOI: 10.1016/B978-0-7020-4226-3.00010-X.
26. Jackson P, Blythe D. Immunohistochemical techniques. Suvarna SK, Layton C, Bancroft JD, editors. Theory & Practice of Histological Techniques. 7th ed. ch 18. Philadelphia: Churchill Livingstone;2013. p. 382–434.
Article
27. Moller DE. New drug targets for type 2 diabetes and the metabolic syndrome. Nature. 2001; 414:821–827. DOI: 10.1038/414821a. PMID: 11742415.
Article
28. Nashwa MT, Heba SS, Laila AR, Amal EF. Role of vitamin D and metformine on pancreatic tissue in an experimental model of type II diabetes mellitus. International Archives of BioMedical and Clinical Research. 2016; 2:3–10.
29. Fraulob JC, Ogg-Diamantino R, Fernandes-Santos C, Aguila MB, Mandarim-de-Lacerda CA. A mouse model of metabolic syndrome: insulin resistance, fatty liver and non-alcoholic fatty pancreas disease (NAFPD) in C57BL/6 mice fed a high fat diet. J Clin Biochem Nutr. 2010; 46:212–223. DOI: 10.3164/jcbn.09-83. PMID: 20490316. PMCID: 2872226.
Article
30. Robertson RP. Chronic oxidative stress as a central mechanism for glucose toxicity in pancreatic islet beta cells in diabetes. J Biol Chem. 2004; 279:42351–42354. DOI: 10.1074/jbc.R400019200. PMID: 15258147.
Article
31. Sattar N, Gill JM. Type 2 diabetes as a disease of ectopic fat? BMC Med. 2014; 12:123–129. DOI: 10.1186/s12916-014-0123-4. PMID: 25159817. PMCID: 4143560.
Article
32. Kluth O, Mirhashemi F, Scherneck S, Kaiser D, Kluge R, Neschen S, Joost HG, Schürmann A. Dissociation of lipotoxicity and glucotoxicity in a mouse model of obesity associated diabetes: role of forkhead box O1 (FOXO1) in glucose-induced beta cell failure. Diabetologia. 2011; 54:605–616. DOI: 10.1007/s00125-010-1973-8. PMCID: 3034032.
Article
33. Song MY, Bae UJ, Lee BH, Kwon KB, Seo EA, Park SJ, Kim MS, Song HJ, Kwon KS, Park JW, Ryu DG, Park BH. Nardostachys jatamansi extract protects against cytokine-induced beta-cell damage and streptozotocin-induced diabetes. World J Gastroenterol. 2010; 16:3249–3257. DOI: 10.3748/wjg.v16.i26.3249. PMID: 20614480. PMCID: 2900716.
Article
34. Ezquer F, Ezquer M, Contador D, Ricca M, Simon V, Conget P. The antidiabetic effect of mesenchymal stem cells is unrelated to their transdifferentiation potential but to their capability to restore Th1/Th2 balance and to modify the pancreatic microenvironment. Stem Cells. 2012; 30:1664–1674. DOI: 10.1002/stem.1132. PMID: 22644660.
Article
35. Baragli A, Grande C, Gesmundo I, Settanni F, Taliano M, Gallo D, Gargantini E, Ghigo E, Granata R. Obestatin enhances in vitro generation of pancreatic islets through regulation of developmental pathways. PLoS One. 2013; 8:e64374. DOI: 10.1371/journal.pone.0064374. PMID: 23741322. PMCID: 3669302.
Article
36. al-Shamaony L, al-Khazraji SM, Twaij HA. Hypoglycaemic effect of Artemisia herba alba. II. Effect of a valuable extract on some blood parameters in diabetic animals. J Ethnopharmacol. 1994; 43:167–171. DOI: 10.1016/0378-8741(94)90038-8. PMID: 7990489.
Article
37. Ahmed D, Sharma M, Mukerjee A, Ramteke PW, Kumar V. Improved glycemic control, pancreas protective and hepatoprotective effect by traditional poly-herbal formulation “Qurs Tabasheer” in streptozotocin induced diabetic rats. BMC Complement Altern Med. 2013; 13:10–25. DOI: 10.1186/1472-6882-13-10.
Article
38. Agnew A, Calderwood D, Chevallier OP, Greer B, Grieve DJ, Green BD. Chronic treatment with a stable obestatin analog significantly alters plasma triglyceride levels but fails to influence food intake; fluid intake; body weight; or body composition in rats. Peptides. 2011; 32:755–762. DOI: 10.1016/j.peptides.2010.12.005.
Article
39. Smitka K, Papezova H, Vondra K, Hill M, Hainer V, Nedvidkova J. The role of “mixed” orexigenic and anorexigenic signals and autoantibodies reacting with appetite-regulating neuropeptides and peptides of the adipose tissue-gut-brain axis: relevance to food intake and nutritional status in patients with anorexia nervosa and bulimia nervosa. Int J Endocrinol. 2013; DOI: 10.1155/2013/483145.
Article
40. Xu G, Kaneto H, Laybutt DR, Duvivier-Kali VF, Trivedi N, Suzuma K, King GL, Weir GC, Bonner-Weir S. Downregulation of GLP-1 and GIP receptor expression by hyperglycemia: possible contribution to impaired incretin effects in diabetes. Diabetes. 2007; 56:1551–1558. DOI: 10.2337/db06-1033. PMID: 17360984.
41. Holz GG. Epac: A new cAMP-binding protein in support of glucagon-like peptide-1 receptor-mediated signal transduction in the pancreatic beta-cell. Diabetes. 2004; 53:5–13. DOI: 10.2337/diabetes.53.1.5.
Article
42. Buteau J. GLP-1 receptor signaling: effects on pancreatic beta-cell proliferation and survival. Diabetes Metab. 2008; 34(Suppl 2):S73–S77. DOI: 10.1016/S1262-3636(08)73398-6. PMID: 18640589.
43. Butler PC, Meier JJ, Butler AE, Bhushan A. The replication of beta cells in normal physiology, in disease and for therapy. Nat Clin Pract Endocrinol Metab. 2007; 3:758–768. DOI: 10.1038/ncpendmet0647. PMID: 17955017.
Article
44. Yang BT, Dayeh TA, Volkov PA, Kirkpatrick CL, Malmgren S, Jing X, Renström E, Wollheim CB, Nitert MD, Ling C. Increased DNA methylation and decreased expression of PDX-1 in pancreatic islets from patients with type 2 diabetes. Mol Endocrinol. 2012; 26:1203–1212. DOI: 10.1210/me.2012-1004. PMID: 22570331. PMCID: 5416998.
Article
45. Vishwakarma SK, Rahamathulla S, Bardia A, Tiwari SK, Srinivas G, Raj A, Tripura C, Sandhya A, Habeeb MA, Khan AA, Pande G, Reddy KP, Reddy PY. In vitro quantitative and relative gene expression analysis of pancreatic transcription factors Pdx-1, Ngn-3, Isl-1, Pax-4, Pax-6 and Nkx-6.1 in trans-differentiated human hepatic progenitors. J Diabetes Investig. 2014; 5:492–500. DOI: 10.1111/jdi.12193. PMID: 25411615. PMCID: 4188105.
Article
46. Granata R, Ghigo E. Products of the ghrelin gene, the pancreatic β-cell and the adipocyte. Endocr Dev. 2013; 25:144–156. DOI: 10.1159/000346306.
Article
47. Bansal VS, Raja CP, Venkataraman K, Vijayalakshmi MA. Genes involved in pancreatic islet cell rejuvenation. Indian J Med Res. 2013; 137:695–703. PMID: 23703336. PMCID: 3724249.
48. Wassef MA, Fouad H, Sabry D, Afifi N, Abbas AM, Mostafa W, Ahmed SH. Therapeutic efficacy of differentiated versus undifferentiated mesenchymal stem cells in experimental type I diabetes in rat. Biochem Biophys Rep. 2016; 5:468–475. PMID: 28955854. PMCID: 5600460.
Article
49. Bonner-Weir S, Li WC, Ouziel-Yahalom L, Guo L, Weir GC, Sharma A. Beta-cell growth and regeneration: replication is only part of the story. Diabetes. 2010; 59:2340–2348. DOI: 10.2337/db10-0084. PMID: 20876724. PMCID: 3279552.
Full Text Links
  • IJSC
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr