Abstract

Aims/hypothesisThe aim of this study was to examine the effects of proinflammatory cytokines on cells of different developmental stages during the generation of stem cell-derived beta cells (SC-beta cells) from human pluripotent stem cells (hPSCs). We wanted to find out to what extent human SC-beta cells are suitable as an experimental cellular model and, with regard to a possible therapeutic use, whether SC-beta cells have a comparable vulnerability to cytokines as bona fide beta cells.MethodshPSCs were differentiated towards pancreatic organoids (SC-organoids) using a 3D production protocol. SC-beta cells and non-insulin-producing cells were separated by FACS and differential gene expression profiles of purified human SC-beta cells, progenitor stages and the human beta cell line EndoC-βH1, as a reference, were determined after 24 h incubation with the proinflammatory cytokines IL-1β, TNF-α and IFN-γ via a transcriptome microarray. Furthermore, we investigated apoptosis based on caspase cleavage, the generation of reactive oxygen species and activation of mitogen-activated protein-kinase (MAPK) stress-signalling pathways.ResultsA 24 h exposure of SC-beta cells to proinflammatory cytokines resulted in significant activation of caspase 3/7 and apoptosis via the extrinsic and intrinsic apoptosis signalling pathways. At this time point, SC-beta cells showed a markedly higher sensitivity towards proinflammatory cytokines than non-insulin-producing cells and EndoC-βH1 cells. Furthermore, we were able to demonstrate the generation of reactive oxygen species and rule out the involvement of NO-mediated stress. A transient activation of stress-signalling pathways p38 mitogen-activated protein kinases (p38) and c-Jun N-terminal kinase (JNK) was already observed after 10 min of cytokine exposure. The transcriptome analysis revealed that the cellular response to proinflammatory cytokines increased with the degree of differentiation of the cells. Cytokines induced the expression of multiple inflammatory mediators including IL-32, CXCL9 and CXCL10 in SC-beta cells and in non-insulin-producing cells.Conclusions/interpretationOur results indicate that human SC-beta cells respond to proinflammatory cytokines very similarly to human islets. Due to the fast and fulminant cellular response of SC-beta cells, we conclude that SC-beta cells represent a suitable model for diabetes research. In light of the immaturity of SC-beta cells, they may be an attractive model for developmentally young beta cells as they are, for example, present in patients with early-onset type 1 diabetes. The secretion of chemotactic signals may promote communication between SC-beta cells and immune cells, and non-insulin-producing cells possibly participate in the overall immune response and are thus capable of amplifying the immune response and further stimulating inflammation. We demonstrated that cytokine-treated SC-organoids secrete IL-32, which is considered a promising candidate for type 1 diabetes onset. This underlines the need to ensure the survival of SC-beta cells in an autoimmune environment such as that found in type 1 diabetes.Graphical abstract

Highlights

  • Type 1 diabetes is a chronic autoimmune disease characterised by loss of insulin production resulting from the selective and progressive destruction of beta cells

  • To determine the responsiveness of the cells to the proinflammatory cytokines IL-1β, TNF-α and IFN-γ, we first analysed the expression of their corresponding receptors during the course of differentiation towards stem cell-derived organoids (SC-organoids) in comparison with the widely used human EndoC-βH1 beta cell line [25]

  • The strong expression of IL1 receptor 1 (IL1R1) in purified SC-beta cells is of particular interest, since human islet beta cells are known to be sensitive to IL-1β-mediated beta cell destruction [27,28,29,30]

Read more

Summary

Introduction

Type 1 diabetes is a chronic autoimmune disease characterised by loss of insulin production resulting from the selective and progressive destruction of beta cells. Activated immune cells infiltrate the islets of Langerhans and mediate their harmful effects through soluble cytotoxic mediators and cell–cell contacts. Proinflammatory cytokines, such as IL-1β, TNF-α and IFN-γ, are considered to be major mediators for the destruction of beta cells [1,2,3]. ER stress, IL-1 and/or TNF can induce the stress-signalling pathways c-Jun Nterminal kinase (JNK) and p38 mitogen-activated protein kinases (p38) [5,6,7]. Cytokine exposure induces expression of MHC class I and II proteins and secretion of cytokines and chemokines such as chemokine (C-X-C motif) ligand 9/10 (CXCL9/10), CC-chemokine ligand 2/3/5/ 8 (CCL2/3/5/8) and others by beta cells, thereby promoting a destructive dialogue between immune cells and beta cells, resulting in acceleration of islet inflammation (insulitis) [8, 9]

Objectives
Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call