open access

Vol 81, No 4 (2022)
Original article
Submitted: 2021-07-05
Accepted: 2021-09-09
Published online: 2021-09-15
Get Citation

The role of the adipose tissue-derived mesenchymal stem cells enriched with melatonin in pancreatic cellular regeneration

H. El Aasar1, L. Rashed2, A. El Sadik13, R. Amer1, H. Emam1
·
Pubmed: 34545556
·
Folia Morphol 2022;81(4):931-941.
Affiliations
  1. Department of Anatomy and Embryology, Faculty of Medicine, Cairo University, Cairo, Egypt
  2. Department of Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Cairo, Egypt
  3. Department of Anatomy and Histology, College of Medicine, Qassim University, Buraidah, Saudi Arabia

open access

Vol 81, No 4 (2022)
ORIGINAL ARTICLES
Submitted: 2021-07-05
Accepted: 2021-09-09
Published online: 2021-09-15

Abstract

Background: Adipose tissue-derived mesenchymal stem cells (AD-MSCs) were proved to differentiate into insulin-producing cells (IPCs), but the amount of insulin secreted was relatively low compared to the insulin secreted by mature pancreatic islets. Enrichment of MSCs culture with melatonin (MT) was found to promote cartilage matrix synthesis, osteogenic and neuronal differentiation. Therefore, the present study was conducted to evaluate the potential role of MT pre-treated AD-MSCs in enhancing the treatment and regeneration of the islet cells of Langerhans in rats with diabetes induced by streptozotocin (STZ).
Materials and methods: Forty adult male Sprague Dawley albino rats were divided equally into groups; group I (control group), group II (STZ group), group III (STZ + AD-MSCs) and group IV (STZ+MT pre-treated AD-MSCs). Biochemical studies were implemented including measurements of the body weight, fasting blood glucose and serum insulin levels, Interleukin 17 (IL-17) and IL-10. Samples of the pancreas were taken and prepared for light, fluorescent microscopic examination, proliferating cell nuclear antigen and caspase-3 immunohistochemical studies and histomorphometric analysis.
Results: The present study confirmed the regenerative and therapeutic effects of AD-MSCs on the pancreatic cells. Concomitant supply of MT to the culture of AD-MSCs, in group IV, was shown to retain the normal architecture of the islet cells of Langerhans. They appeared well-defined and lightly stained, surrounded by classical pancreatic acini and contained a large number of islet cells with vesicular nuclei and prominent nucleoli. Improvement of all the biochemical parameters, in the same group, was demonstrated by increased body weight and serum insulin levels with a decrease in the fasting blood glucose levels. Significant decrease in the pro-inflammatory cytokine; IL-17 and increase in the anti-inflammatory cytokine; IL-10, compared to the STZ group, were also discovered. Significant increase in the proliferating cell nuclear antigen proliferation index, decrease in caspase-3 and increase in PKH26 labelled MSCs area per cent was recorded in the group of AD-MSCs enriched with MT compared to the group of AD-MSCs without MT.
Conclusions: The present study confirmed the potential therapeutic and protective role of MT pre-treated AD-MSCs against the STZ-induced pancreatic islet cells damage. Further studies are recommended to investigate the efficacy of MT and AD-MSCs over longer experimental durations.

Abstract

Background: Adipose tissue-derived mesenchymal stem cells (AD-MSCs) were proved to differentiate into insulin-producing cells (IPCs), but the amount of insulin secreted was relatively low compared to the insulin secreted by mature pancreatic islets. Enrichment of MSCs culture with melatonin (MT) was found to promote cartilage matrix synthesis, osteogenic and neuronal differentiation. Therefore, the present study was conducted to evaluate the potential role of MT pre-treated AD-MSCs in enhancing the treatment and regeneration of the islet cells of Langerhans in rats with diabetes induced by streptozotocin (STZ).
Materials and methods: Forty adult male Sprague Dawley albino rats were divided equally into groups; group I (control group), group II (STZ group), group III (STZ + AD-MSCs) and group IV (STZ+MT pre-treated AD-MSCs). Biochemical studies were implemented including measurements of the body weight, fasting blood glucose and serum insulin levels, Interleukin 17 (IL-17) and IL-10. Samples of the pancreas were taken and prepared for light, fluorescent microscopic examination, proliferating cell nuclear antigen and caspase-3 immunohistochemical studies and histomorphometric analysis.
Results: The present study confirmed the regenerative and therapeutic effects of AD-MSCs on the pancreatic cells. Concomitant supply of MT to the culture of AD-MSCs, in group IV, was shown to retain the normal architecture of the islet cells of Langerhans. They appeared well-defined and lightly stained, surrounded by classical pancreatic acini and contained a large number of islet cells with vesicular nuclei and prominent nucleoli. Improvement of all the biochemical parameters, in the same group, was demonstrated by increased body weight and serum insulin levels with a decrease in the fasting blood glucose levels. Significant decrease in the pro-inflammatory cytokine; IL-17 and increase in the anti-inflammatory cytokine; IL-10, compared to the STZ group, were also discovered. Significant increase in the proliferating cell nuclear antigen proliferation index, decrease in caspase-3 and increase in PKH26 labelled MSCs area per cent was recorded in the group of AD-MSCs enriched with MT compared to the group of AD-MSCs without MT.
Conclusions: The present study confirmed the potential therapeutic and protective role of MT pre-treated AD-MSCs against the STZ-induced pancreatic islet cells damage. Further studies are recommended to investigate the efficacy of MT and AD-MSCs over longer experimental durations.

Get Citation

Keywords

melatonin, adipose tissue-derived mesenchymal stem cells, islets of Langerhans, diabetes; interleukin 17, interleukin 10

About this article
Title

The role of the adipose tissue-derived mesenchymal stem cells enriched with melatonin in pancreatic cellular regeneration

Journal

Folia Morphologica

Issue

Vol 81, No 4 (2022)

Article type

Original article

Pages

931-941

Published online

2021-09-15

Page views

4423

Article views/downloads

881

DOI

10.5603/FM.a2021.0093

Pubmed

34545556

Bibliographic record

Folia Morphol 2022;81(4):931-941.

Keywords

melatonin
adipose tissue-derived mesenchymal stem cells
islets of Langerhans
diabetes
interleukin 17
interleukin 10

Authors

H. El Aasar
L. Rashed
A. El Sadik
R. Amer
H. Emam

References (28)
  1. Ammar HI, Sequiera GL, Nashed MB, et al. Comparison of adipose tissue- and bone marrow- derived mesenchymal stem cells for alleviating doxorubicin-induced cardiac dysfunction in diabetic rats. Stem Cell Res Ther. 2015; 6: 148.
  2. Basoli V, Santaniello S, Cruciani S, et al. Melatonin and vitamin D interfere with the adipogenic fate of adipose-derived stem cells. Int J Mol Sci. 2017; 18(5).
  3. Berebichez-Fridman R, Montero-Olvera PR. Sources and clinical applications of mesenchymal stem cells: state-of-the-art review. Sultan Qaboos Univ Med J. 2018; 18(3): e264–e277.
  4. Bloom DD, Centanni JM, Bhatia N, et al. A reproducible immunopotency assay to measure mesenchymal stromal cell-mediated T-cell suppression. Cytotherapy. 2015; 17(2): 140–151.
  5. Chandra V, Swetha G, Muthyala S, et al. Islet-like cell aggregates generated from human adipose tissue derived stem cells ameliorate experimental diabetes in mice. PLoS One. 2011; 6(6): e20615.
  6. Cho J, D'Antuono M, Glicksman M, et al. A review of clinical trials: mesenchymal stem cell transplant therapy in type 1 and type 2 diabetes mellitus. Am J Stem Cells. 2018; 7(4): 82–93.
  7. El-Said KS, Ezz AA, El Ba, et al. The potential role of male bone marrow mesenchymal stem cells of diabetic female rats. Diabetes Manag. 2018; 8(6): 137–146.
  8. Hashemian SJ, Kouhnavard M, Nasli-Esfahani E. Mesenchymal stem cells: rising concerns over their application in treatment of type one diabetes mellitus. J Diabetes Res. 2015; 2015: 675103.
  9. Hu C, Li L. Melatonin plays critical role in mesenchymal stem cell-based regenerative medicine in vitro and in vivo. Stem Cell Res Ther. 2019; 10(1): 13.
  10. Huang L, Wang G. The effects of different factors on the behavior of neural stem cells. Stem Cells Int. 2017; 2017: 9497325.
  11. Karaoz E, Okcu A, Ünal ZS, et al. Adipose tissue-derived mesenchymal stromal cells efficiently differentiate into insulin-producing cells in pancreatic islet microenvironment both in vitro and in vivo. Cytotherapy. 2013; 15(5): 557–570.
  12. Luchetti F, Canonico B, Bartolini D, et al. Melatonin regulates mesenchymal stem cell differentiation: a review. J Pineal Res. 2014; 56(4): 382–397.
  13. Mortezaee K, Khanlarkhani N, Sabbaghziarani F, et al. Preconditioning with melatonin improves therapeutic outcomes of bone marrow-derived mesenchymal stem cells in targeting liver fibrosis induced by CCl4. Cell Tissue Res. 2017; 369(2): 303–312.
  14. Moshtagh PR, Emami SH, Sharifi AM. Differentiation of human adipose-derived mesenchymal stem cell into insulin-producing cells: an in vitro study. J Physiol Biochem. 2013; 69(3): 451–458.
  15. Nawaz M, Fatima F, Vallabhaneni KC, et al. Extracellular vesicles: evolving factors in stem cell biology. Stem Cells Int. 2016; 2016: 1073140.
  16. Omar A, Aboulkhair A. Effect of bone marrow versus adipose tissue derived mesenchymal stem cells on the pancreas of streptozotocin-induced diabetes mellitus type i in adult male rats (histological study). Egypt J Histol. 2017; 40(1): 12–24.
  17. Pei M, He F, Wei L, et al. Melatonin enhances cartilage matrix synthesis by porcine articular chondrocytes. J Pineal Res. 2009; 46(2): 181–187.
  18. Sneddon JB, Tang Q, Stock P, et al. Stem cell therapies for treating diabetes: progress and remaining challenges. Cell Stem Cell. 2018; 22(6): 810–823.
  19. Stehle JH, Saade A, Rawashdeh O, et al. A survey of molecular details in the human pineal gland in the light of phylogeny, structure, function and chronobiological diseases. J Pineal Res. 2011; 51(1): 17–43.
  20. Teo GSL, Ankrum JA, Martinelli R, et al. Mesenchymal stem cells transmigrate between and directly through tumor necrosis factor-α-activated endothelial cells via both leukocyte-like and novel mechanisms. Stem Cells. 2012; 30(11): 2472–2486.
  21. Thakkar UG, Trivedi HL, Vanikar AV, et al. Insulin-secreting adipose-derived mesenchymal stromal cells with bone marrow-derived hematopoietic stem cells from autologous and allogenic sources for type 1 diabetes mellitus. Cytotherapy. 2015; 17(7): 940–947.
  22. Ullah M, Liu DD, Thakor AS. Mesenchymal stromal cell homing: mechanisms and strategies for improvement. iScience. 2019; 15: 421–438.
  23. Volarevic V, Al-Qahtani A, Arsenijevic N, et al. Interleukin-1 receptor antagonist (IL-1Ra) and IL-1Ra producing mesenchymal stem cells as modulators of diabetogenesis. Autoimmunity. 2010; 43(4): 255–263.
  24. Wang Y, Chen X, Cao W, et al. Plasticity of mesenchymal stem cells in immunomodulation: pathological and therapeutic implications. Nat Immunol. 2014; 15(11): 1009–1016.
  25. Zang Li, Hao H, Liu J, et al. Mesenchymal stem cell therapy in type 2 diabetes mellitus. Diabetol Metab Syndr. 2017; 9: 36.
  26. Zhang HT, Liu ZL, Yao XQ, et al. Neural differentiation ability of mesenchymal stromal cells from bone marrow and adipose tissue: a comparative study. Cytotherapy. 2012; 14(10): 1203–1214.
  27. Zhang Y, Ge XH, Guo XJ, et al. Bone marrow mesenchymal stem cells inhibit the function of dendritic cells by secreting galectin-1. Biomed Res Int. 2017; 2017: 3248605.
  28. Zhao L, Hu C, Zhang P, et al. Melatonin preconditioning is an effective strategy for mesenchymal stem cell-based therapy for kidney disease. J Cell Mol Med. 2020; 24(1): 25–33.

Regulations

Important: This website uses cookies. More >>

The cookies allow us to identify your computer and find out details about your last visit. They remembering whether you've visited the site before, so that you remain logged in - or to help us work out how many new website visitors we get each month. Most internet browsers accept cookies automatically, but you can change the settings of your browser to erase cookies or prevent automatic acceptance if you prefer.

By VM Media Group sp. z o.o., Grupa Via Medica, Świętokrzyska 73, 80–180 Gdańsk, Poland

tel.: +48 58 320 94 94, faks: +48 58 320 94 60, e-mail: viamedica@viamedica.pl