Abstract

The genetic factors that underlie the increasing incidence of diabetes with age are poorly understood. We examined whether telomere length, which is inherited and known to shorten with age, plays a role in the age-dependent increased incidence of diabetes. We show that in mice with short telomeres, insulin secretion is impaired and leads to glucose intolerance despite the presence of an intact β-cell mass. In ex vivo studies, short telomeres induced cell-autonomous defects in β-cells including reduced mitochondrial membrane hyperpolarization and Ca2+ influx which limited insulin release. To examine the mechanism, we looked for evidence of apoptosis but found no baseline increase in β-cells with short telomeres. However, there was evidence of all the hallmarks of senescence including slower proliferation of β-cells and accumulation of p16INK4a. Specifically, we identified gene expression changes in pathways which are essential for Ca2+-mediated exocytosis. We also show that telomere length is additive to the damaging effect of endoplasmic reticulum stress which occurs in the late stages of type 2 diabetes. This additive effect manifests as more severe hyperglycemia in Akita mice with short telomeres which had a profound loss of β-cell mass and increased β-cell apoptosis. Our data indicate that short telomeres can affect β-cell metabolism even in the presence of intact β-cell number, thus identifying a novel mechanism of telomere-mediated disease. They implicate telomere length as a determinant of β-cell function and diabetes pathogenesis.

Highlights

  • The incidence of type 2 diabetes increases with age and one in five individuals are affected by the age of 60 [1]

  • Mice with short telomeres have impaired glucose tolerance and glucose-stimulated insulin release To test whether short telomeres impair glucose homeostasis, we studied late generation CAST/EiJ mice that are heterozygous null for telomerase RNA, mTR+/2, and have short telomeres (Figure S1A)

  • The study of the telomerase knockout mouse has particular significance for understanding the genetics of type 2 diabetes since the short telomere defect that is present in this model is acquired universally in human aging

Read more

Summary

Introduction

The incidence of type 2 diabetes increases with age and one in five individuals are affected by the age of 60 [1]. Type 2 diabetes manifests as impaired glucose tolerance and defective insulin secretion which occur in the presence of an intact b-cell mass [2]. This is followed by a frank loss of b-cells and a concomitant need for insulin therapy [2,3,4]. Recent genomic studies have underscored the influence of inherited factors that affect b-cell integrity and function in age-related diabetes [5,6,7,8] While these germline factors are hypothesized to play a role in the pathogenesis of diabetes, the biology that underlies their increasing penetrance with age is not understood

Methods
Results
Conclusion
Full Text
Paper version not known

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

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.