Abstract

Dysfunctional islets of Langerhans are a hallmark of type 2 diabetes (T2D). We hypothesize that differences in islet gene expression alternative splicing which can contribute to altered protein function also participate in islet dysfunction. RNA sequencing (RNAseq) data from islets of obese diabetes-resistant and diabetes-susceptible mice were analyzed for alternative splicing and its putative genetic and epigenetic modulators. We focused on the expression levels of chromatin modifiers and SNPs in regulatory sequences. We identified alternative splicing events in islets of diabetes-susceptible mice amongst others in genes linked to insulin secretion, endocytosis or ubiquitin-mediated proteolysis pathways. The expression pattern of 54 histones and chromatin modifiers, which may modulate splicing, were markedly downregulated in islets of diabetic animals. Furthermore, diabetes-susceptible mice carry SNPs in RNA-binding protein motifs and in splice sites potentially responsible for alternative splicing events. They also exhibit a larger exon skipping rate, e.g., in the diabetes gene Abcc8, which might affect protein function. Expression of the neuronal splicing factor Srrm4 which mediates inclusion of microexons in mRNA transcripts was markedly lower in islets of diabetes-prone compared to diabetes-resistant mice, correlating with a preferential skipping of SRRM4 target exons. The repression of Srrm4 expression is presumably mediated via a higher expression of miR-326-3p and miR-3547-3p in islets of diabetic mice. Thus, our study suggests that an altered splicing pattern in islets of diabetes-susceptible mice may contribute to an elevated T2D risk.

Highlights

  • More than 420 million people worldwide suffer from type 2 diabetes (T2D), meaning that 1 out of 11 adults is affected, and the prevalence is constantly rising

  • New Zealand Obese (NZO) and B6-ob/ob-mice differ in their susceptibility to developing diabetes

  • RNA sequencing (RNAseq) data from isolated islets of both strains were screened for alternative splicing events with a focus on (i) differential expression of histones and chromatin modifiers, (ii) microexons, (iii) enriched RNA binding protein motifs, and (iv) SNPs located in splicing relevant sequences (Figure 1A)

Read more

Summary

Introduction

More than 420 million people worldwide suffer from type 2 diabetes (T2D), meaning that 1 out of 11 adults is affected, and the prevalence is constantly rising. Pancreatic β-cells can compensate for insulin resistance by increasing β-cell mass and insulin secretion. Obese mice which are hyperglycemic and insulin-resistant, such as New Zealand Obese (NZO) mice, are used as a model system in this context. B6-ob/ob mice, which carry a mutation in the Leptin gene, become obese and insulin-resistant without developing diabetes, and serve as non-diabetic controls. Working with mouse models of divergent genotypes and different diabetes susceptibility gives insights into genetic marks that are associated with the onset and progression of the disease. Transcriptome analysis of the two mouse strains identified significant differences

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