AuPS Logo Programme
Contents
Previous Next PDF

RCAN1 is critical for β-cell mitochondrial function and is overexpressed in human Type 2 diabetes

H. Peiris,1 M. Duffield,1 C.F. Jessup,2,3 V. Kashmir,1 J. Fadista,4 A. Genders,5 S. McGee,5 M. Ryan,6 D.R. Laybutt,7 P.T. Coates,8 M.A. Pritchard,9 L. Groop4 and D.J. Keating,1 1Molecular and Cellular Neuroscience Group, Department of Human Physiology and Centre for Neuroscience, Flinders University, Adelaide, SA 5042, Australia, 2Islet Biology Laboratory, Department of Anatomy and Histology and Centre for Neuroscience, Flinders University, Adelaide, SA 5042, Australia, 3Discipline of Medicine, University of Adelaide, Adelaide, SA 5001, Australia, 4Lund University Diabetes Centre, Sweden, 5Metabolic Remodelling Laboratory, Metabolic Research Unit, School of Medicine, Deakin University, Geelong, VIC 3220, Australia, 6Mitochondria Laboratory, Department of Biochemistry and Molecular Biology, Monash University, VIC 3800, Australia, 7Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Sydney, NSW Australia, 8Royal Adelaide Hospital, North Terrace, Adelaide, SA 5000, Australia and 9Department of Biochemistry and Molecular Biology, Monash University, VIC 3800, Australia.

Type 2 diabetes (T2D) is a complex metabolic disorder characterized by elevated blood glucose levels. Pancreatic β-cell dysfunction and reduced insulin output in the presence of insulin resistance is the primary defect resulting in T2D. It is unknown what triggers the switch from β-cell compensation to β-cell failure but studies from human T2D islets indicate reduced glucose-stimulated insulin secretion underlined by mitochondrial dysfunction and a diminished capacity to produce ATP. We have established that Regulator of calcineurin 1 (RCAN1) is a stress-induced protein expressed in β-cells that can regulate insulin secretion. We find that RCAN1 expression is increased in T2D islets in humans and mice. Patch clamp studies confirmed that β-cells overexpressing RCAN1 display significant reductions in glucose-induced membrane depolarization and in depolarization-induced exocytosis, with both of these defects being attributable to reduced ATP generation. Inclusion of equimolar ATP inside single β-cells rescued this exocytosis defect. Direct measurements of β-cell mitochondrial output demonstrate that increased RCAN1 perturbs mitochondrial function, and thus ATP production, and that this occurs due to altered functionality of the ADP/ATP translocator. Thus, we provide evidence that increased RCAN1 expression may drive some of the key metabolic changes that underlie the development of β-cell dysfunction in T2D.