Journal of the Pancreas Open Access

  • ISSN: 1590-8577
  • Journal h-index: 80
  • Journal CiteScore: 29.12
  • Journal Impact Factor: 19.45*
  • Average acceptance to publication time (5-7 days)
  • Average article processing time (30-45 days) Less than 5 volumes 30 days
    8 - 9 volumes 40 days
    10 and more volumes 45 days
Reach us +44 7460731551

Rapid Communication - (2022) Volume 23, Issue 12

Signaling Mechanism that Encourages Insulin Secretion from Pancreatic Nestin-Positive Progenitor Cells
Darla M. Domionius*
 
Developmental Biology & Stem Cell Research, Hubrecht Institute, Utrecht, The Netherlands
 
*Correspondence: Darla M. Domionius, Developmental Biology & Stem Cell Research, Hubrecht Institute, Utrecht, The Netherlands, Email:

Received: 28-Nov-2022 Editor assigned: 29-Nov-2022 Reviewed: 09-Dec-2022 Revised: 14-Dec-2022 Published: 21-Dec-2022, DOI: 10.35841/1590-8577- 23.12.780

Abstract

The failure of the beta-cells in the pancreatic islets of Langerhans to produce enough insulin to meet the body's needs results in the condition known as diabetes mellitus. In patients with advanced diabetes, whole pancreas or islet transplants successfully restore insulin production. The development of alternate sources of islets, such as ex vivo culture and differentiation of stem/progenitor cells, is necessary due to the shortage of donor pancreata. Previously, it was identified nestin-positive islet-derived progenitor cells (NIPs) as multipotent progenitor cells in adult human pancreatic islets that express the neural stem cell marker nestin. The NIP cells, which are also found in the pancreatic ducts, could play a role in where islet progenitor cells are found. The discovery of NIP cells within the pancreatic islets itself raises the possibility of treating diabetes by expanding NIP cells isolated from pancreas biopsies outside of the recipient or donor body.

Introduction

Islet cell transplantation may be a useful strategy for type 1 diabetes cell replacement. There aren't many islets available for allogeneic transplantation, though. Developing strategies to halt the course of type 2 diabetes may be made easier by understanding the formation of new -cells in adulthood. The intermediate filament protein nestin is well-known as a neuroepithelial stem cell marker. Within the pancreas, nestin-positive cells are likely a population of progenitor cells. Both neuronal and islet cell types may have a common precursor stem cell marker called nestin. This system has the potential to be a new source of -cells and can be used to explore the mechanism of islet neogenesis, making it easier to find drug targets to treat insulin-dependent diabetes. Retinoic acid was employed to encourage pancreatic differentiation in the first step (chromatin remodelling), which is consistent with the earlier finding that RA is necessary for early pancreas development in humans and mice. The failure of the islets of Langerhans to produce insulin leads to the development of the disease diabetes mellitus [1]. In patients with advanced diabetes, whole pancreas or islet transplants successfully restore insulin production. The creation of alternate sources of islets is necessary since donor pancreata are not always available. Efforts to produce insulin-secreting beta cells from a variety of progenitor populations have been sparked by the promising outcomes of pancreatic islet transplantation for the treatment of diabetes mellitus and the acute shortage of donor pancreata [2]. In this paper, we provide a novel culture technique for obtaining insulinproducing cells from rat bone marrow cells that are nestinpositive (n-BMSC). With the use of our method, n-BMSC were successfully made to express transcription factors specific to various lineages, including Pdx1, which has been demonstrated to promote pancreatic differentiation. The production of PDX1-positive pancreatic progenitor cells from mouse embryonic stem cells has been actively encouraged by the application of all-trans RA. In our investigation, RA was employed to encourage pancreatic phenotypic differentiation of n-BMSC. It was successfully encouraged to differentiate into a more mature phenotype that included ductal and insulin-producing cells. Among numerous tissues, including the pancreas, nestin is a neuroepithelial marker that is briefly produced in the early phases [3]. We demonstrated how altering n-BMSC can gradually increase the expression of transcription factors involved in early pancreatic and endocrine specification. It was successfully encouraged to differentiate into a more mature phenotype that included ductal and insulinproducing cells. For diabetic patients, islet transplantation is a promising way to replenish their functional islet cell mass. There are only so many human donor islets available, hence it is essential to investigate innovative methods as potential renewable alternatives to transplantation. Largescale proliferation and the ability to differentiate into multiple lineages are characteristics of stem cells [4]. They could provide crucial information for cell replacement therapy. Human embryonic stem cells (hESCs) can spontaneously differentiate into cells that make insulin (IPCs). Additionally, by altering the culture medium's composition and expressing the major transcription factors involved in pancreas formation, significant success has recently been made in getting ESCs to preferentially differentiate into pancreatic lineages [5].

Conclusion

Insulin-producing cells IPCs made using the nestin and DE procedures closely resemble real insulin-secreting cells. However, compared to adult human islets, the expression levels of markers specific to pancreatic islets are substantially lower. Despite this, there are still many obstacles to be solved before either protocol can be used to produce high-quality PCs on a big scale.

References

  1. Bouwens L, Kloppel G. Islet cell neogenesis in the pancreas. Virchows Archiv 1996; 427:553-560. [PMID: 8605565].
  2. Indexed at, Google Scholar, Cross Ref

  3. Alpert S, Hanahan D, Teitelman G. Hybrid insulin genes reveal a developmental lineage for pancreatic endocrine cells and imply a relationship with neurons. Cell 1988; 53(2):295-308. [PMID: 3282675].
  4. Indexed at, Google Scholar, Cross Ref

  5. Zulewski H, Abraham EJ, Gerlach MJ, Daniel PB, Moritz W, Muller B, et al. Multipotential nestin-positive stem cells isolated from adult pancreatic islets differentiate ex vivo into pancreatic endocrine, exocrine, and hepatic phenotypes. Diabetes 2001;50(3):521-33. [PMID: 11246871].
  6. Indexed at, Google Scholar, Cross Ref

  7. 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;100(3):998-1003. [PMID: 12525695].
  8. Indexed at, Google Scholar, Cross Ref

  9. Wei R, Yang J, Hou W, Liu G, Gao M, Zhang L, et al. Insulin-producing cells derived from human embryonic stem cells: comparison of definitive endoderm-and nestin-positive progenitor-based differentiation strategies. PloS one 2013;8(8):e72513. [PMID: 23951327].
  10. Indexed at, Google Scholar, Cross Ref

Copyright: 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 work is properly cited.