[HTML][HTML] Inhibition of Miro1 disturbs mitophagy and pancreatic β-cell function interfering insulin release via IRS-Akt-Foxo1 in diabetes

L Chen, C Liu, J Gao, Z Xie, LWC Chan, DJ Keating… - Oncotarget, 2017 - ncbi.nlm.nih.gov
L Chen, C Liu, J Gao, Z Xie, LWC Chan, DJ Keating, Y Yang, J Sun, F Zhou, Y Wei, X Men…
Oncotarget, 2017ncbi.nlm.nih.gov
Mitochondrial function is essential to meet metabolic demand of pancreatic beta cells
respond to high nutrient stress. Mitophagy is an essential component to normal pancreatic β-
cell function and has been associated with β-cell failure in Type 2 diabetes (T2D). Our
previous studies have indicated that mitochondrial Rho (Miro) GTPase-mediated
mitochondrial dysfunction under high nutrient stress leads to NOD-like receptor 3 (NLRP3)-
dependent proinflammatory responses and subsequent insulin resistance. However, the in …
Abstract
Mitochondrial function is essential to meet metabolic demand of pancreatic beta cells respond to high nutrient stress. Mitophagy is an essential component to normal pancreatic β-cell function and has been associated with β-cell failure in Type 2 diabetes (T2D). Our previous studies have indicated that mitochondrial Rho (Miro) GTPase-mediated mitochondrial dysfunction under high nutrient stress leads to NOD-like receptor 3 (NLRP3)-dependent proinflammatory responses and subsequent insulin resistance. However, the in vivo mechanism by which Miro1 underlies mitophagy has not been identified. Here we show firstly that the expression of Miro is reduced in human T2D and mouse db/db islets and in INS-1 cell line exposed to high glucose and palmitate. β-cell specific ablation of Miro1 (Miro1f/f: Rip-cre mice, or (IKO) under high nutrient stress promotes the development of hyperglycemia. β-cells from IKO mice display an inhibition of mitophagy under oxidative stress and induces mitochondrial dysfunction. Dysfunctional mitophagy in IKO mice is represented by damaged islet beta cell mitochondrial and secretory capacity, unbalanced downstream MKK-JNK signalling without affecting the levels of MEK, ERK or p38 activation and subsequently, impaired insulin secretion signaling via inhibition IRS-AKT-Foxo1 pathway, leading to worsening glucose tolerance in these mice. Thus, these data suggest that Miro1 may be responsible for mitophagy deficiency and β-cell dysfunction in T2D and that strategies target Miro1 in vivo may provide a therapeutic target to enhance β-cell mitochondrial quality and insulin secretion to ameliorate complications associated with T2D.
ncbi.nlm.nih.gov