Abstract

Connections between deficient autophagy and insulin resistance have emerged, however, the mechanism through which reduced autophagy impairs insulin-signaling remains unknown. We examined mouse embryonic fibroblasts lacking Atg16l1 (ATG16L1 KO mouse embryonic fibroblasts (MEFs)), an essential autophagy gene, and observed deficient insulin and insulin-like growth factor-1 signaling. ATG16L1 KO MEFs displayed reduced protein content of insulin receptor substrate-1 (IRS1), pivotal to insulin signaling, whereas IRS1myc overexpression recovered downstream insulin signaling. Endogenous IRS1 protein content and insulin signaling were restored in ATG16L1 KO mouse embryonic fibroblasts (MEF) upon proteasome inhibition. Through proximity-dependent biotin identification (BioID) and co-immunoprecipitation, we found that Kelch-like proteins KLHL9 and KLHL13, which together form an E3 ubiquitin (Ub) ligase complex with cullin 3 (CUL3), are novel IRS1 interactors. Expression of Klhl9 and Klhl13 was elevated in ATG16L1 KO MEFs and siRNA-mediated knockdown of Klhl9, Klhl13, or Cul3 recovered IRS1 expression. Moreover, Klhl13 and Cul3 knockdown increased insulin signaling. Notably, adipose tissue of high-fat fed mice displayed lower Atg16l1 mRNA expression and IRS1 protein content, and adipose tissue KLHL13 and CUL3 expression positively correlated to body mass index in humans. We propose that ATG16L1 deficiency evokes insulin resistance through induction of Klhl9 and Klhl13, which, in complex with Cul3, promote proteasomal IRS1 degradation.

Highlights

  • Connections between deficient autophagy and insulin resistance have emerged, the mechanism through which reduced autophagy impairs insulin-signaling remains unknown

  • Diminished actin dynamics contributes to insulin resistance additional to defects in AKT activity [8, 9, 26, 27] and might be an element in whole-body insulin resistance observed in models of deficient autophagy

  • The various signaling defects observed in ATG16L1 KO mouse embryonic fibroblasts (MEF) could arise at a number of levels within the insulin signaling cascade

Read more

Summary

The abbreviations used are

T2D, type 2 diabetes; IGF1, insulin-like growth factor-1; Ub, ubiquitin; ATG16L1, autophagy-related 16L1; KLHL, Kelchlike; IRS1, insulin receptor substrate 1; BioID, proximity-dependent biotin identification; IR, insulin receptor; CUL3, Cullin 3; LC3, microtubule-associated light chain 3B; PI3K, phosphoinositide 3-kinase; PI[4,5]P2, phosphatidylinositol 4,5-bisphosphate; PI[3,4,5]P3, phosphatidylinositol [3,4,5]-trisphosphate; PH, pleckstrin homology; PDK1, phosphoinositide-dependent kinase 1; SOCS, suppressor of cytokine signaling; HFD, high-fat diet; LFD, low-fat diet; BMI, body mass index; PMSF, phenylmethylsulfonyl fluoride; TPCK, L-1-tosylamido-2-phenylethyl chloromethyl ketone; ER, endoplasmic reticulum; KO, knock-out; MEF, mouse embryonic fibroblast; FBS, fetal bovine serum; DMEM, Dulbecco’s modified Eagle’s medium; qPCR, quantitative PCR; ANOVA, analysis of variance; JNK, c-Jun N-terminal kinase; GAPDH, glyceraldehyde-3-phosphate dehydrogenase; GSK3␤, glycogen synthase kinase 3␤. One proposed mechanism for the obesity-linked reduction in IRS1 invokes inflammation and endoplasmic reticulum (ER) stress [10, 11] This leads to IRS1 serine-phosphorylation, and ubiquitination via a suppressor of cytokine signaling 1 (SOCS1)/SOCS3-containing E3 ubiquitin (Ub) ligase complex that targets IRS1 for proteasomal degradation [7, 12,13,14]. We show that ATG16L1 deficiency causes insulin resistance in MEFs and propose a responsible mechanism implicating the KLHL9/KLHL13/CUL3 E3 Ub ligase complex in IRS1 degradation. These findings highlight a novel pathway leading to targeted degradation of IRS1 and causally to subsequent downstream defects in insulin signaling

Results
Discussion
Experimental procedures
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